18.01.2013 Views

Editorial Cover Story ESOP/NZW 2011 Congress Report Oncology ...

Editorial Cover Story ESOP/NZW 2011 Congress Report Oncology ...

Editorial Cover Story ESOP/NZW 2011 Congress Report Oncology ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Editorial</strong><br />

Hope is a good breakfast but a bad supper 5<br />

ECOP www.esop.eu<br />

European Conference of<br />

<strong>Oncology</strong> Pharmacy<br />

<strong>Cover</strong> <strong>Story</strong><br />

<strong>ESOP</strong>/<strong>NZW</strong> <strong>2011</strong> <strong>Congress</strong> <strong>Report</strong><br />

Preparation of monoclonal antibodies: practice across 6<br />

Europe<br />

The role of a pharmacist according to patients 10<br />

Companion diagnostics and personalised cancer medicine 12<br />

An assessment of the pharmacist’s role in oncology trial 14<br />

management<br />

Breast cancer irradiation: <strong>2011</strong> update 16<br />

Extravasation of cytotoxics: there is still room for 18<br />

improvement<br />

<strong>Oncology</strong> Pharmacy Practice<br />

DNA repair in cancer therapy: beneficial or harmful 21<br />

Cold water reconstitution of Vidaza® with subsequent 24<br />

refrigerated storage prolongs drug stability<br />

A practical approach to oral tumour therapy 26<br />

Setting up a pharmacist-led chemotherapy clinic: a 28<br />

practical guide<br />

Update<br />

Clinical evaluation of lenograstim use in the Bank of 31<br />

Cyprus <strong>Oncology</strong> Centre<br />

Special <strong>Report</strong><br />

Hospital workers perceptions about nanotechnology 35<br />

Announcement 34<br />

EJOP is published quarterly and mailed to more than 5,000 European oncology pharmacists, pharmacy<br />

technicians and subscribers in 33 countries; and distributed at major international and national conferences.<br />

EJOP is available online (www.ejop.eu).<br />

Guest Authors:<br />

Dr Mirjam Crul, Dr Ann-Sophie Franki, Dr Kathleen Simons,Anita Waldmann, Dr Jan Trøst Jørgensen, Maria<br />

Hersom, Dr Mikael Daouphars, Ewelina Korczowska, Dr Martin Sevec, Professor Dr Wolfgang Wagner,<br />

Dr Ursula Pluschnig, Shahla Farokhnia, Gunar Stemer, Professor Robert M Mader, Professor Dorothee C<br />

Dartsch, Dr Anthony Tutino, Dr Mei Lai, Jürgen Barth, Carl Booth,Andri Kouroufexi, Stavroula Theophanous-<br />

Kitiri, George Polykarpou, Dr Yiola Markou, Dr Evangelia Papadimitriou, Dr Nagwa Ibrahim, Dr Ali Al Zahrani,<br />

Dr Ashraf Al Alwan<br />

EJOP <strong>Editorial</strong> Office:<br />

Postbus 10001, BE-2400 Mol, Belgium<br />

Tel.: +32 474 989572<br />

Fax: +32 14 583048<br />

editorial@ppme.eu - www.ejop.eu<br />

EJOP <strong>Editorial</strong> Board:<br />

Dr Robert Terkola,Austria<br />

Professor Alain Astier, France<br />

Professor Dr Wolfgang Wagner, Germany<br />

Professor Dr Günther Wiedemann, Germany<br />

Professor Per Hartvig, Sweden<br />

Publisher:<br />

Lasia Tang - Ltang@ejop.eu<br />

Editor-in-Chief:<br />

Klaus Meier - kmeier@ejop.eu<br />

European Journal of <strong>Oncology</strong> Pharmacy<br />

Senior Executive Editor:<br />

Esra Kurt, PhD - ek@ppme.eu<br />

Editors:<br />

Neil Goodman, PhD - ng@ppme.eu<br />

Steve Dawber, BSc (Hons) - editorial@ppme.eu<br />

Marketing Assistant/Subscriptions:<br />

Jodi Honea - info@ppme.eu<br />

Production Assistant:<br />

Rachel Mortishire-Smith - support@ppme.eu<br />

Science Assistant:<br />

Gaynor Ward - science@ppme.eu<br />

ISSN EJOP: 1783-3914<br />

Print Run: 5,000 Printed by PPS s.a.<br />

Published in Belgium<br />

by Pharma Publishing &<br />

Media Europe<br />

copyright@ppme.eu<br />

EJOP • Volume 5 • <strong>2011</strong> • Issues 3-4 • €35<br />

www.ejop.eu<br />

Correspondents:<br />

Austria: Dr Robert Terkola/Vienna<br />

Belgium: Isabelle Glorieux/Wilrijk<br />

Croatia: Vesna Pavlica/Zagreb<br />

Cyprus: Stavroula Kitiri/Nikosia<br />

Czech Republic: Irena Netikova/Prague<br />

Denmark: Eva Honoré/Copenhagen<br />

Estonia: Kristjan Kongi/Tallinn<br />

Finland:Wilppu Terhi/Turku<br />

France: Professor Alain Astier/Paris<br />

Germany: Dr Michael Höckel/Eisenach<br />

Greece: Ioanna Saratsiotou/Athens<br />

Hungary: Mónika Kis Szölgyémi/Budapest<br />

Iceland:Thorir Benediktssson/Reykjavík<br />

Italy: Franca Goffredo/Turin<br />

Lithuania: Birutė Varanavičienė/Vilnius<br />

Luxembourg: Camille Groos/Luxembourg<br />

Malta: Fiona Grech/La Valetta<br />

Netherlands: Kathleen Simons/Nijmegen<br />

Poland: Dr Jerzy Lazowski/Warsaw<br />

Portugal: Margarida Caramona/Coimbra<br />

Serbia and Montenegro:Tatjana Tomic/Belgrade<br />

Slovak Republic: Maria Karpatova/Bratislava<br />

Slovenia: Monika Sonc/Ljubljana<br />

Spain: Dr Maria José Tamés/San Sebastian<br />

Sweden: Professor Per Hartvig-Honoré/Uppsala<br />

Switzerland: Monique Ackermann/Geneva<br />

United Kingdom: Jeff Koundakijan/Wales<br />

Subscription Rates 2012:<br />

Europe Non-Europe<br />

Individual: €135 €159<br />

Hospital/University: €297 €321<br />

Corporate: €378 €402<br />

Student: € 81 €105<br />

Individual and student subscriptions are subject<br />

to 21% VAT Belgian Government tax.<br />

COPYRIGHT<br />

© <strong>2011</strong> Pharma Publishing and Media Europe.All rights reserved. Materials printed in EJOP are covered by copyright, throughout<br />

the world and in all languages.The reproduction, transmission or storage in any form or by any means either mechanical<br />

or electronic, including subscriptions, photocopying, recording or through an information storage and retrieval system of the<br />

whole, or parts of the, article in these publications is prohibited without consent of the publisher.The publisher retains the<br />

right to republish all contributions and submitted materials via the Internet and other media.<br />

Individuals may make single photocopies for personal non-commercial use without obtaining prior permission.Non-profit institutions<br />

such as hospitals are generally permitted to make copies of articles for research or teaching activities (including multiple<br />

copies for classrooms use) at no charge, provided the institution obtains the prior consent from Pharma Publishing and<br />

Media Europe. For more information contact the publisher.<br />

DISCLAIMER<br />

While the publisher, editor(s) and editorial board have taken every care with regard to accuracy of editorial and advertisement<br />

contributions, they cannot be held responsible or in any way liable for any errors, omissions or inaccuracies contained therein<br />

or for any consequences arising therefrom.<br />

Statements and opinions expressed in the articles and communications herein are those of the author(s) and do not necessarily<br />

reflect the views of the publisher, editor(s) and editorial board.<br />

Neither the publisher nor the editor(s) nor the editorial board guarantees,warrants,or endorses any product or service advertised<br />

in this publication, nor do they guarantee any claim made by the manufacturer of such product or service. Because of<br />

rapid advances in the medical sciences, in particular, independent verification of diagnoses and drug dosages should be made.<br />

SUBSCRIPTIONS<br />

EJOP is published quarterly. Subscription orders must be prepaid. Subscriptions paid are non-refundable. Contact<br />

marketing@ppme.eu for further information.<br />

Change of address notices: both subscriber’s old and new addresses should be sent to marketing@ppme.eu at least one<br />

month in advance.<br />

Claims: when claims for undelivered or damaged issues are made within four months of publication of the issue, a<br />

complimentary replacement issue will be provided.


The discussion about the future<br />

role of pharmacy is ongoing and<br />

at the European level several proposals<br />

from ministries or the<br />

pharmaceutical inspection have<br />

been tendered along the way to describe the<br />

possible needs. We know that several lobbying<br />

groups are working in Strasbourg, France,<br />

as well as in Brussels, Belgium, to bring about<br />

different points of view about the position of<br />

pharmacists.<br />

Unnoticed by the public, the path is being<br />

smoothed to ignore the resolution adopted by the<br />

Council of Europe Committee of Ministers in<br />

1997 [1]:<br />

- Recalling that the pharmacist’s evolving role is to ensure the<br />

safe and effective use of medicines by patients at the lowest cost,<br />

thus contributing to the welfare of the patients and the improvement<br />

of public health;<br />

- Considering the present trend to shift the focus of attention away<br />

from the preparation of medicines towards ensuring that they<br />

are appropriately prescribed and dispensed by pharmacists and<br />

correctly used by patients;<br />

- Considering that this trend requires legislative reform and<br />

changes in initial training, and makes ethical demands on the<br />

profession;<br />

- Considering that, in a changing economic and social environment,<br />

pharmacists must adapt and must bring up to date their<br />

knowledge, training and practice in order to play their full part<br />

in society;<br />

- Considering that the patient’s well-being must be at the centre of<br />

all pharmaceutical activity, that the pharmacist’s traditional<br />

focus on the medicine must be shifted to the patient, and that the<br />

pharmacist should become a provider of health care in which the<br />

patient occupies the central place …<br />

The understanding of the role of pharmacists is and was clear.<br />

Near to patients in collaboration with doctors, defining the multiprofessional<br />

work and improving their knowledge and expertise.<br />

Since that time the <strong>ESOP</strong> has developed four new editions of<br />

Quality Standard for the <strong>Oncology</strong> Pharmacy Service (QuapoS),<br />

whilst keeping in close contact with the European Commissioner<br />

for Health and Consumer Policy. The goal is to unify national<br />

conditions for preparing cytotoxics and to educate healthcare<br />

workers as well as patients. This was no preterm birth that could<br />

not be followed by pharmacists Europe-wide. It is an open guide-<br />

<strong>Editorial</strong><br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

Hope is a good breakfast but a bad supper *<br />

Klaus Meier<br />

Editor-in-Chief<br />

line with an option for each country to<br />

enshrine it in national legislation.<br />

<strong>ESOP</strong>’s understanding has grown that we<br />

have to foster collaboration with the European<br />

CanCer Organisation (ECCO) because a joint<br />

platform insures a multi-professional<br />

approach and the opportunity to be recognised<br />

as full partners of healthcare teams. Apharmacist<br />

will again be present on the board of<br />

directors in the next legislative period.<br />

EJOP has now been published for five years<br />

and it plays its part in harmonising and<br />

encouraging the process of oncology pharmacy<br />

in Europe at the same time.<br />

Again, in this issue we are presenting the efforts of national work<br />

in the report about the preparation of monoclonal antibodies as well<br />

as the general role of pharmacists in oncology trial management.<br />

We get surprising views about the conditions for pharmacists<br />

from some countries. We can learn how in the UK the chemotherapy<br />

clinics can be led by pharmacists and follow the discussion<br />

from Saudi Arabia about hospital workers’ understanding of nanotechnology.<br />

Often work at the national level triggers a European<br />

discussion. So, a questionnaire will be sent to all our 2,600<br />

members about their understanding of nanotechnology in order to<br />

sensitise them to both the benefits and the risks.<br />

As history has taught us that the revolution devours its children,<br />

we are aware we must show our responsibility every day in our<br />

daily work in each country. We are proud to celebrate in January<br />

2012 the 20th <strong>NZW</strong>—clinical conference for oncology pharmacy<br />

departments—with nearly 1,000 participants and are sure that<br />

the first European Conference of <strong>Oncology</strong> Pharmacy in<br />

Budapest, Hungary in September 2012 will give all European<br />

oncology pharmacists a place of exchange and mutual support.<br />

We are able and willing to work the whole day in not only the<br />

hope but also in the solid conviction that pharmacists, as we<br />

understand ourselves, are needed and welcome.<br />

I am pleased and proud to be a president of such committed and<br />

convinced membership.<br />

*Sir Francis Bacon<br />

Reference<br />

1. Council of Europe Committee of Ministers [homepage on the Internet].<br />

Resolution AP (97) 2 on the development of the function of pharmacists<br />

and the adaptation of their initial training. [cited <strong>2011</strong> Dec 15]. Available<br />

from: wcd.coe.int/ViewDoc.jsp?id=596013&Site=CM<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

5


<strong>Cover</strong> <strong>Story</strong> - <strong>ESOP</strong>/<strong>NZW</strong> <strong>2011</strong> <strong>Congress</strong> <strong>Report</strong><br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

Preparation of monoclonal antibodies: practice<br />

across Europe<br />

To ensure proper handling of monoclonal antibodies (mAbs), the hospital pharmacist has to consider the safety of<br />

the product, the safety of the patient, and the safety of the personnel compounding the drugs. We conducted a survey<br />

across Europe to investigate current practice.<br />

Introduction<br />

Ever since the first monoclonal<br />

antibodies (mAbs) became available,<br />

hospital pharmacists have<br />

been faced with the challenge of<br />

preparing them so that patients<br />

receive the correct dose, infusions<br />

are sterile, and pharmacy<br />

staff are protected from potentially<br />

harmful exposure. Formal<br />

studies investigating the risk of<br />

mAb-related occupational expo-<br />

Mirjam Crul<br />

PharmD, PhD<br />

sure have not yet been performed. Hence, when compounding<br />

parenteral formulations of these drugs in the hospital setting,<br />

the responsible pharmacist must subjectively assess the magnitude<br />

of risk. This raises several issues. Firstly, given that a<br />

large proportion of mAbs are administered to immunologically<br />

challenged patients, and sometimes with concomitant<br />

chemotherapy, a thorough aseptic technique which minimises<br />

the risk of microbiological contamination is pivotal.<br />

Secondly, most mAbs are dosed using body weight or body<br />

surface area. Therefore, a safe and validated method of calculating<br />

the correct amount of infusion concentrate to add to the<br />

diluent is required.<br />

Thirdly, occupational exposure of pharmacy and nursing personnel<br />

should be considered. Although most mAbs do not have a direct<br />

effect on DNA and are therefore not mutagenic or genotoxic, therapeutic<br />

exposure to some mAbs, e.g. muromonab, has been linked<br />

to tumour development [1], possibly via indirect pathways. Given<br />

that most hospital staff are only exposed to trace amounts of mAbs,<br />

this risk is likely to be very small. However, because evidence<br />

exists to show that rituximab, trastuzumab, and antiangiogenic<br />

mAbs such as bevacizumab, cetuximab and panitumumab can<br />

harm foetal development [2-5], handling guidelines are needed for<br />

pregnant or lactating hospital staff. Furthermore, because all mAbs<br />

are proteins they may cause sensitivity reactions. As yet, there is no<br />

way of establishing safe concentrations or MAC values of monoclonal<br />

antibodies for healthcare workers. Fortunately, when compared<br />

with classical chemotherapeutics, more stringent endogenous<br />

barriers exist against mAbs. Monoclonal antibodies have a<br />

relatively large molecular weight (about 150 kDa) and in the<br />

absence of special techniques such as microneedles or electroporation,<br />

are unable to pass the skin barrier [6]. Additionally,<br />

Monoclonal antibodies are extremely susceptible to denaturation<br />

induced by pH changes or gastrointestinal enzymes. Hence, oral<br />

Ann-Sophie Franki<br />

PharmD, PhD<br />

Kathleen Simons<br />

PharmD<br />

absorption of these molecules is<br />

highly unlikely, but the risk of pulmonary<br />

absorption of mAB-containing<br />

aerosols, e.g. those created<br />

during vial reconstitution using<br />

syringes and needles, cannot be<br />

fully excluded [7].<br />

Finally, one needs to consider<br />

whether it is safe to use the same<br />

equipment to prepare mAbs and<br />

cytostatics. In theory, it is possi-<br />

ble that a safety cabinet or isolator could become contaminated<br />

with traces of chemotherapeutics [8-10] and if, for example,<br />

an infliximab infusion is prepared following a cytostatic compounding<br />

session, it may become cross-contaminated. Because<br />

drugs such as infliximab are generally administered on nononcological<br />

wards, this contamination may then spread to<br />

other hospital departments where personal protection measures<br />

are not as strict as those of the oncology ward.<br />

Risk assessment<br />

In The Netherlands, a mAb risk classification is currently<br />

being executed. Each drug is assessed using a standardised<br />

method that has been developed by an independent Dutch<br />

research institute (TNO, Dutch Institute for Applied Sciences),<br />

which includes an assessment of the drug’s carcinogenicity,<br />

teratogenicity, and other toxic properties. Each drug is classified<br />

in one of five groups, with the highest class 5 being the<br />

most hazardous and class 1 being considered harmless.<br />

Cytostatics often fall into class 5. To assess toxic effects,<br />

exposure limits, material safety data sheets, and preclinical<br />

and clinical studies are reviewed. Using this method, mAbs<br />

generally fall into class 3 or 4.<br />

Under Dutch national recommendations, compounding in a<br />

safety cabinet or isolator is recommended for class 4 compounds,<br />

whereas compounds regarded as class 3 require only<br />

the provision of personal protection (protective clothing and<br />

gloves). Thus far, five of the eighteen investigated mAbs have<br />

been deemed class 4 (alemtuzumab, bevacizumab, cetuximab,<br />

muromonab, panitumomab) and one (ibritumomab) has even<br />

been deemed class 5, based on its radioactivity. Further mAb<br />

classifications are listed in Table 1.<br />

In Germany, a risk assessment of the most frequently used<br />

mAbs has been published [11]. After a thorough investigation<br />

6 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


Table 1: Risk assessment of monoclonal antibodies performed in The Netherlands<br />

Name Indication Chimeric-human Possible risk Conclusion - class<br />

Abciximab Trombocytaggregation inhibition Chimeric No 3<br />

Adalimumab Rheumatoid arthritis, Crohn’s disease Human Not likely 3<br />

Alemtuzumab Leukaemia Human Reprotoxicity, serious 4<br />

immuno suppression<br />

Basiliximab Organ transplants Chimeric Not likely 3<br />

Bevacizumab Colorectal/mamma carcinoma Human Embryotoxic 4<br />

Cetuximab Colorectal carcinoma Chimeric Embryotoxic 4<br />

Ibritumomab Non-Hodgkin’s lymphoma Human Radioactive 5<br />

Infliximab Rheumatoid arthritis, Crohn’s disease Chimeric Not likely 3<br />

Muromonab Organ transplants Human Carcinogenic 4<br />

Natalizumab Multiple sclerosis Human Not likely 3<br />

Omalizumab Asthma Human No 3<br />

Palivizumab Respiratory syncytial virus Human No 3<br />

Panitumumab Colorectal carcinoma Human Embryotoxic, decreased 4<br />

fertility<br />

Rituximab Non-Hodgkin’s lymphoma, Chimeric Not likely 3<br />

rheumatoid arthritits<br />

Trastuzumab Mamma carcinoma Human Not likely 3<br />

of all available safety data, the study concluded that mAbs<br />

should be handled as hazardous substances and prepared in<br />

safety cabinets using maximum personal protection—as<br />

described in German national regulations.<br />

Survey of practices across Europe<br />

To help us define a national guideline, we conducted an email<br />

survey amongst <strong>ESOP</strong> delegates. Thirty countries were<br />

approached, and completed questionnaires were received from<br />

20 country representatives: Belgium (2 respondents), Bosnia,<br />

Croatia, Cyprus, Czech Republic, Denmark, Estonia (2<br />

respondents), Finland, France, Germany, Hungary, Italy,<br />

Malta, Poland, Portugal, Slovenia, Spain, Switzerland, The<br />

Netherlands, Turkey. Two countries, Belgium and Estonia,<br />

provided two separate replies, resulting in a total of 22 completed<br />

questionnaires.<br />

The first question asked was ‘where and by whom in the hospital<br />

were mAbs prepared?’ In 17 cases (77%), they were prepared in<br />

the pharmacy, in four cases (18%) they were prepared on the wards<br />

by nurses, and in 0 cases (0%) they were prepared on the wards by<br />

pharmacy staff. In some instances, however, mAbs for chemotherapy<br />

patients were prepared in the pharmacy, but mAbs for all other<br />

patient types were prepared on the wards, see Figure 1.<br />

The second question asked was ‘what type of equipment was used<br />

in the preparation of mAbs?’ The vast majority of respondents (n<br />

= 15, 66%) used a safety cabinet. Five respondents (22%) reported<br />

using an isolator, and three respondents (14%) said no equipment<br />

or a crossflow cabinet was used, see Figure 2.<br />

The next set of questions concerned the use of separate equipment<br />

for mAb preparation in non-chemotherapy patients. We<br />

asked these questions to review the possibility of cross-contamination,<br />

e.g. traces of chemotherapy residues on infusions for<br />

patients with rheumatoid arthritis or Crohn’s disease. Because<br />

many <strong>ESOP</strong> members work in dedicated chemotherapy clinics,<br />

only nine respondents were able to answer these questions. Five<br />

respondents used a separate safety cabinet or isolator, and four<br />

respondents reported that mAbs and cytotoxics were prepared<br />

using the same equipment in non-chemotherapy patients.<br />

Respondents were also asked whether their compounding equipment<br />

was cleaned between mAb and chemotherapeutic use. Six<br />

respondents reported that such a procedure existed in their hospital,<br />

but 12 respondents reported that this was not the case. The<br />

other four respondents said this question was not applicable in<br />

their situation, mainly because they had separate equipment for<br />

these distinct procedures.<br />

The final question asked was ‘what type of transfer device was<br />

used when preparing mAbs?’, see Figure 3. Responses to this<br />

question were diverse. Five respondents used only needles,<br />

four respondents used only spikes, and three respondents used<br />

closed transfer systems. However, most respondents (n = 7)<br />

reported that they used a combination of these devices.<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

7


Figure 1: Where mAbs are prepared<br />

in your hospital<br />

<strong>Cover</strong> <strong>Story</strong> - <strong>ESOP</strong>/<strong>NZW</strong> <strong>2011</strong> <strong>Congress</strong> <strong>Report</strong><br />

The results of our survey provided a clear impression of the<br />

variety of practice across Europe. In this respect, it was striking<br />

that in countries where two different hospitals completed<br />

the questionnaire, the responses also differed. This suggests<br />

that, in keeping with practice in The Netherlands, none of the<br />

surveyed countries have a broad consensus or guideline on this<br />

topic.<br />

Recommendations and QuapoS guidelines<br />

In 2008, an <strong>ESOP</strong> technical workshop was held in Hamburg,<br />

Germany, and the subject of preparing mAbs, chemotherapy,<br />

active vaccines, and gene medicine was discussed. This workshop<br />

provided some recommendations on the issue:<br />

• Cytotoxics and mAbs for cancer patients can be prepared<br />

using the same equipment.<br />

• Monoclonal antibodies for non-cancer patients that have<br />

cytotoxic, mutagenic or reprotoxic (teratogenic) activities<br />

(CMR), should be prepared using safety equipment (safety<br />

cabinet or isolator) but in a different one than that which is<br />

used for the preparation of chemotherapy. If this is not possible,<br />

an appropriate cleaning procedure before and after the<br />

preparation must be established.<br />

These recommendations are due to be incorporated in the next<br />

edition of the Quality Standard for the <strong>Oncology</strong> Pharmacy<br />

Service with Commentary (QuapoS). As a result of the Dutch<br />

risk assessment, we believe that this draft <strong>ESOP</strong> guidance is<br />

valid, but only for approximately one-third of mAbs, i.e. the ones<br />

that we believe have CMR activities, or that are suspected of<br />

being cytotoxic or teratogenic. In these cases, one should prepare<br />

the drugs in a safety cabinet or isolator, using spikes or closed<br />

devices, in line with the compounding practice for cytotoxics.<br />

Authors<br />

Mirjam Crul, PharmD, PhD<br />

Hospital Pharmacist<br />

Division of Clinical Pharmacy<br />

Onze Lieve Vrouwe Gasthuis<br />

9 Oosterpark<br />

NL-1090 HM Amsterdam, The Netherlands<br />

Figure 2: Type of equipment used<br />

when preparing mAbs<br />

Ann-Sophie Franki, PharmD, PhD<br />

Central Pharmacy<br />

Universitair Ziekenhuis Gent<br />

185 De Pintelaan<br />

BE-9000 Gent, Belgium<br />

Kathleen Simons, PharmD<br />

Hospital Pharmacist<br />

Division of Clinical Pharmacy<br />

Sint Elizabeth Ziekenhuis<br />

60 Hilvarenbeekse Weg<br />

NL-5022 GC Tilburg, The Netherlands<br />

Figure 3: Type of transfer device used<br />

when preparing mAbs<br />

mAbs: monoclonal antibodies; Grey dots represent countries that were approached through the <strong>ESOP</strong> forum but did not return a completed questionnaire.<br />

References<br />

1. Orthoclone OKT3, Summary of product characteristics. Janssen-<br />

Cilag, 2010.<br />

2. NIOSH. List of new FDA drugs and warnings fitting NIOSH criteria<br />

for hazardous drugs 2006. Available from: www.cdc.gov/niosh/docket/<br />

archive/docket105.html<br />

3. Halsen G, Kramer I. Gefahrdungsbeurteilung monoklonaler antikorper<br />

der ATC klasse L01XC. Krankenhauspharmazie. 2009;30:441-53.<br />

4. Chakravarty EF, Murray ER, Kelman A, Farmer P. Pregnancy outcomes<br />

following maternal exposure to rituximab. Blood. <strong>2011</strong>;117 (5):1499-506.<br />

5. Shrim A, Garcia-Bournissen F, Maxwell C, Farine D, Koren G.<br />

Trastuzumab treatment for breast cancer during pregnancy. Can Fam<br />

Physician. 2008;54(1):31-2.<br />

6. Mitragotri, S. Immunization without needles. Nat Rev Immunol.<br />

2005;5(12):905-16.<br />

7. Wang W, Wang EQ, Balthasar JP. Monoclonal antibody pharmacokinetics<br />

and pharmacodynamics. Clin Pharmacol Ther. 2008;84(5):548-58.<br />

8. Sessink PJ, Anzion RB, Van den Broek PH, Bos RP. Detection of<br />

contamination with antineoplastic agents in a hospital pharmacy<br />

department. Pharm Weekbl Sci.1992;14(1):16-22.<br />

9. Schulz H, Bigelow S, Dobish R, Chambers CR. Antineoplastic agent<br />

workplace contamination study: the Alberta Cancer Board Pharmacy<br />

perspective. J Oncol Pharm Pract. 2005;11(3):101-9.<br />

10. Mason HJ, Blair S, Sams C, et al. Exposure to antineoplastic drugs in two<br />

UK hospital pharmacy units. Ann Occup Hyg. 2005;49(7):603-10.<br />

11. Halsen G, Kramer I. Forschung BGW. Bewertung monoklonaler anti-korper<br />

zum schutz beschaftiger. 30 Oktober 2008. Available from: www.bgwonline.de<br />

bestellnummer EP- BmAk<br />

8 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


<strong>Cover</strong> <strong>Story</strong> - <strong>ESOP</strong>/<strong>NZW</strong> <strong>2011</strong> <strong>Congress</strong> <strong>Report</strong><br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

The role of a pharmacist according to patients<br />

From a patient’s point of view, pharmacists play a key role in the healthcare team through the provision of highquality<br />

medication, appropriate information on treatment schedules and effects of medication, as well as advice on<br />

the management of adverse events and use of complementary treatments.<br />

Introduction<br />

In recent years, substantial changes have been<br />

made to our approaches to healthcare provision.<br />

An increasing emphasis has been placed<br />

on a multidisciplinary approach to care, as<br />

well as treatment in the community setting. In<br />

addition, a substantial evolution in the way<br />

patients are involved in the management of their<br />

diseases (patient participation) has taken place.<br />

Many patients are keen to have an active say in<br />

their treatment and often look for additional<br />

material to supplement the information offered by their healthcare<br />

providers [1]. Indeed, a ‘new consumer’ has been described by<br />

pharmacists, predominantly young customers or the chronically ill,<br />

who are well informed, inquisitive, with a desire for longer<br />

dialogue and who ask critical questions and seek counselling [2].<br />

Accordingly, the objective of patient management strategies has<br />

been defined to provide a patient-centred approach.<br />

The pharmacist plays a key role both as a partner within the healthcare<br />

team, actively involved in the delivery of care through the provision<br />

of high quality medication on the one hand, as well as being<br />

a source of appropriate information and education regarding<br />

treatment schedules, desirable effects, as well as side effects of<br />

medication and including advice on the management of adverse<br />

events. This demonstrates a substantial maturation in the role of the<br />

pharmacist. In the future, pharmacists may also play a greater role<br />

in the independent prescribing of medicines [3].<br />

Overview: patient expectations<br />

Patients look to their pharmacist not as a salesperson, but as a<br />

skilled professional with up-to-date knowledge and expertise,<br />

to be able to offer appropriate advice and information both on<br />

general and specific health issues [4]. Key attributes of a pharmacist<br />

are respecting the privacy and confidentiality of<br />

patients, as well as offering compassion and empathy in discussions<br />

of their diseases, treatments or concerns, which highlights<br />

the importance of good communication and interpersonal<br />

skills [4-6]. Overall, in order to be beneficial, the relationship<br />

between pharmacists and patients needs to be built on<br />

mutual respect and trust.<br />

The results of a survey by Worley et al. which investigated the<br />

views of patient and pharmacist concerning the expectations<br />

both patients and pharmacists have of each other are revealing<br />

in this respect [7]. The survey employed a questionnaire to collect<br />

information on the following aspects: information sharing,<br />

responsible behaviour, interpersonal communication, creating<br />

a patient-centred relationship, and active communication relat-<br />

Anita Waldmann<br />

ed to health care. The study revealed that both<br />

patients and pharmacists have similar views<br />

about the pharmacist’s role in sharing information,<br />

such as how to look out for side effects<br />

and whether a co-administration of other medicines<br />

is feasible. However, patients agreed less on<br />

pharmacists’ responsible behaviour, which<br />

specifically concerned working with patients to<br />

meet their health needs. In addition, patients<br />

agreed less about the pharmacist’s role in creating<br />

a patient-centred relationship and interpersonal<br />

communications, which related to availability and approachability<br />

from a patient’s point of view. Although these responses highlight<br />

aspects of the patient–pharmacist relationship that may benefit<br />

from improvements, the results, which were derived from<br />

national random samples in the US, may not be representative of<br />

the situation in Europe and, in addition, changes are likely to<br />

have occurred since the publication of this study in 2007.<br />

Providing appropriate information and advice<br />

The amount of information following an initial diagnosis of a<br />

disorder can often be overwhelming and confusing for<br />

patients. In addition, the initiation of a new treatment, especially<br />

oral treatments, is accompanied by a host of questions<br />

and a thorough explanation of expected effects is invaluable.<br />

For many patients, the jargon used in package inserts can often<br />

be unclear and unsettling. In addition, patients may be reluctant<br />

about asking the treating physician for clarification. Here,<br />

pharmacists can play a key role in explaining relevant information<br />

and giving advice in plain language. This involves the<br />

clear communication of administration schedules, as well as<br />

effects of the treatment. In addition, pharmacists are able to<br />

provide tools to patients and their families to deal with side<br />

effects. A detailed explanation of possible symptoms and how<br />

these manifest themselves can greatly reassure patients and<br />

caregivers and reduce feelings of panic and powerlessness<br />

when they arise. Pharmacists can provide valuable education<br />

10 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


egarding steps that patients themselves can take to manage<br />

side effects, as well as outlining signals that indicate serious<br />

side effects, which would require further advice and intervention<br />

from healthcare professionals. Provision of such management<br />

information will empower patients, greatly contributing<br />

to a responsible approach to therapy. Assessment of risks and<br />

ensuring their safety is a key concern for patients [5].<br />

In summary, pharmacists play important roles as educators and<br />

counsellors on specific disease and treatments issues. In addition,<br />

there are chronic disorders, such as diabetes, for which<br />

patients’ needs extend beyond advice on specific interventions<br />

to guidance on sustained lifestyle changes, an example that<br />

demonstrates the role of the pharmacist as an essential part of<br />

the interdisciplinary diabetes care team [8].<br />

Providing information regarding complementary<br />

medicines<br />

Many patients are keen to supplement their treatments with complementary<br />

medicines, which they may not always disclose to<br />

their treating physician simply because they are not aware that<br />

negative drug interactions may arise [1]. Here, pharmacists can<br />

play a proactive and crucial role by asking about supplementary<br />

medicines and explaining the benefits, risks, and possible contraindications.<br />

Indeed, a survey conducted among Australian<br />

community pharmacies found that patients expect pharmacists to<br />

advise them on complementary medicines [1].<br />

Providing information regarding patient support<br />

organisations<br />

Following the diagnosis of a serious or terminal illness,<br />

patients and families may feel isolated and confused and may<br />

benefit from contact with patients and caregivers in similar circumstances.<br />

Pharmacists are well placed to provide links or<br />

contact information for patient support organisations or other<br />

sources of information to assist patients and relatives with<br />

meeting other patients.<br />

Explanation of schedules and the importance of<br />

compliance<br />

Medication schedules can be a source of confusion for patients,<br />

particularly if a number of medications have to be taken which<br />

require particular times or circumstances, e.g. fasting versus<br />

administration with food. Pharmacists play a key role in explaining<br />

schedules of administration, thereby reinforcing the information<br />

provided by the treating physician, as well as being in a good<br />

position to stress the importance of adherence to treatment schedules<br />

to maximise the effects of therapy [9].<br />

Non-compliance is recognised to be a frequent problem in the<br />

outpatient setting [10] and a number of factors specifically<br />

contributing to non-compliance with antibiotic therapy have<br />

been identified. These include the cost of drugs, formulation,<br />

rapid improvement of symptoms, forgetfulness, frequent dosing,<br />

complex regimens, side effects and patient beliefs [10].<br />

Non-compliance is associated with serious consequences, such<br />

as patients receiving inappropriate doses of medication, thereby<br />

affecting clinical outcomes. In addition, there are significant<br />

cost implications, not only because of wasted medications,<br />

but due to additional examinations and interventions that<br />

have to be implemented [10].<br />

Through regular contact with patients or their family members,<br />

pharmacists are uniquely placed to discuss and review feasible<br />

and meaningful goals of therapy with patients to meet their<br />

expectations and ensure compliance to treatment.<br />

Conclusion<br />

The role of the pharmacist has matured substantially over<br />

recent years to encompass not only the dispensing of medicine,<br />

but also a substantial involvement in disease management.<br />

Pharmacists play a key role in providing relevant information<br />

and advice on the practical aspects of treatment and are therefore<br />

essential partners in the multidisciplinary teams involved<br />

in the patient-centred provision of healthcare. The relationship<br />

between patients and pharmacists is therefore an integral<br />

aspect of ensuring patient welfare.<br />

Author<br />

Anita Waldmann<br />

Myeloma Euronet AISBL/Leukaemiehilfe Rhein-Main eV<br />

6 Falltorweg<br />

DE-65428 Ruesselsheim, Germany<br />

References<br />

1. Braun LA, Tiralongo E, Wilkinson JM, Spitzer O, Bailey M, Poole S,<br />

et al. Perceptions, use and attitudes of pharmacy customers on complementary<br />

medicines and pharmacy practice. BMC Complement<br />

Altern Med. 2010;10:38.<br />

2. Traulsen JM, Noerreslet M. The new consumer of medicine-the pharmacy<br />

technicians’ perspective. Pharm World Sci. 2004;26(4):203-7.<br />

3. Hobson RJ, Scott J, Sutton J. Pharmacists and nurses as independent<br />

prescribers: exploring the patient’s perspective. Family Practice.<br />

2010;27:110-20.<br />

4. Tinelli M, Bond C, Blenkinsopp A, Jaffray M, Watson M, Hannaford<br />

P; Community Pharmacy Medicines Management Evaluation Team.<br />

Patient evaluation of a community pharmacy medications management<br />

service. Ann Pharmacother. 2007 Dec;41(12):1962-70.<br />

5. Futter B. The naked patient. S Afr Pharm J. <strong>2011</strong>;78(2):56.<br />

6. Naik Panvelkar P, Armour C, Saini B. Community pharmacy-based<br />

asthma services-what do patients prefer? J Asthma. 2010;47(10):<br />

1085-93.<br />

7. Worley MM, Schommer JC, Brown LM, Hadsall RS, Ranelli PL,<br />

Stratton TP, et al. Pharmacists’ and patients’ roles in the pharmacistpatient<br />

relationship: are pharmacists and patients reading from the<br />

same relationship script? Res Social Adm Pharm. 2007;3(1):47-69.<br />

8. Sisson E, Kuhn C. Pharmacist roles in the management of patients<br />

with type 2 diabetes. J Am Pharm Assoc. 2009;49(Suppl 1):S41-5.<br />

9. Vivian EM. The pharmacist’s role in maintaining adherence to insulin therapy<br />

in type 2 diabetes mellitus. Consult Pharm. 2007 Apr;22(4):320-32.<br />

10. Kardas P, Bishai WR. Compliance in anti-infective medicine. Adv<br />

Stud Med. 2006;6(7C):S652-8.<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

11


<strong>Cover</strong> <strong>Story</strong> - <strong>ESOP</strong>/<strong>NZW</strong> <strong>2011</strong> <strong>Congress</strong> <strong>Report</strong><br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

Companion diagnostics and personalised cancer<br />

medicine<br />

Cancer is a heterogeneous disease that requires an individualised treatment approach. Drug treatments guided<br />

by molecular diagnostic testing are increasingly being used in the clinic. This article shows that the concept<br />

of stratified cancer medicine has become a reality.<br />

Introduction<br />

Our increased molecular understanding of<br />

human diseases will change the way that<br />

drugs are developed and how pharmacotherapy<br />

is practiced in the years to come [1, 2].<br />

We know that most diseases are heterogeneous<br />

and that they can be divided into biological<br />

subgroups, each requiring a specific<br />

therapeutic intervention. When it comes to<br />

individualisation of pharmacotherapy, oncol- Jan Trøst Jørgensen<br />

ogy has been at the forefront and a number of MScPharm, PhD<br />

drugs have already been developed based on<br />

a detailed molecular knowledge of the disease mechanisms [3].<br />

Personalised and stratified medicine<br />

When we talk about individualising pharmacotherapy we often use<br />

expressions like ‘tailored therapy’ or ‘personalised medicine’,<br />

which may give the impression of a totally individualised pharmacotherapy<br />

like ‘targeting drugs for each unique genetic profile’ [4].<br />

However, this is yet far from being a reality. What we experience<br />

right now, and will continue to experience much more in the years<br />

to come, is that by using molecular diagnostic tests we will be able<br />

to divide the patients who ‘share’ molecular biological characteris-<br />

Table 1: Drugs for the treatment of solid tumours where companion<br />

diagnostic tests are used<br />

Maria Hersom<br />

BSc<br />

Drug Indication Biomarker<br />

Tamoxifen (Nolvadex, AstraZeneca) Breast cancer ER<br />

Letrozole (Femara, Novartis) Breast cancer ER<br />

Anastrozole (Arimidex, AstraZeneca)<br />

Exemestane (Aromasin, Pfizer)<br />

Trastuzumab (Herceptin, Roche) Breast cancer HER2/HER2<br />

Gastric cancer HER2/HER2<br />

Lapatinib (Tykerb, GlaxoSmithKline) Breast cancer HER2/HER2<br />

Epirubicin (Ellence, Pfizer) Breast cancer TOP2A<br />

Doxorubicin (Adriamycin, Pfizer)<br />

Cetuximab (Erbitux, Colorectal cancer EGFR/KRAS<br />

Bristol-Myers Squibb/Merck)<br />

Panitumumab (Vectibix, Amgen) Colorectal cancer EGFR/KRAS<br />

Gefitinib (Iressa, AstraZeneca) Non-small cell lung EGFR<br />

cancer<br />

Erlotinib (Tarceva, Roche) Non-small cell lung EGFR<br />

cancer<br />

Imatinib (Glivec, Novartis) Gastrointestinal C-KIT (CD117)<br />

stromal tumour<br />

Vemurafenib (Zelboraf, Plexxicon/Roche) Melanoma BRAF V600E<br />

tics into subgroups and, based on this information,<br />

we can match these patients with<br />

the right drug. In this situation, the<br />

molecular diagnostics serve as a stratification<br />

test, which has led to the use of the<br />

expressions ‘stratified medicine’ or ‘stratified<br />

pharmacotherapy’. Stratified medicine<br />

has been defined as follows: management<br />

of a group of patients with<br />

shared biological characteristics by using<br />

molecular diagnostic testing to select the<br />

most optimal therapy in order to achieve<br />

the best possible medicinal outcome for that group [5].<br />

Companion diagnostics<br />

The molecular diagnostics tests used in stratified cancer medicine<br />

rely on different technologies and the dominating analytical methods<br />

are immunohistochemistry (IHC), fluorescence in situ hybridisation<br />

(FISH), chromogenic in situ hybridisation (CISH), and<br />

reverse transcription-polymerase chain reaction (RT-PCR) [3].<br />

When these molecular diagnostics tests are used in combination<br />

with drugs they have been given a specific name. Initially, these<br />

tests were called pharmacodiagnostics or theranostics, but within<br />

the last 3–5 years the expression ‘companion<br />

diagnostics’ has taken over. A companion diagnostics<br />

is defined as: a pre-treatment test performed<br />

in order to determine whether or not a<br />

patient is likely to respond to a given therapy.<br />

This type of test is classified as a predictive test<br />

and a prerequisite for implementation of stratified<br />

and personalised medicine [5].<br />

A number of drugs are listed in Table 1 for the<br />

treatment of solid tumours where the clinical<br />

use is guided by a companion diagnostic test.<br />

For several of these drugs, it is a requirement<br />

that companion diagnostic testing is performed<br />

before they can be prescribed to the patient.<br />

Stratified cancer medicine<br />

Breast cancer, like most other cancers, is a disease<br />

where heterogeneity exists and where the<br />

principle of stratified medicine has in fact been<br />

practised for the last couple of decades. This<br />

already began in the 1970s with the development<br />

of the first selective oestrogen receptor<br />

12 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


modulator tamoxifen. Tamoxifen was developed for women with<br />

oestrogen receptor positive breast cancer, which make up approximately<br />

60–70% of the population with the disease [6]. From a<br />

phase II study with tamoxifen performed in patients with advanced<br />

breast cancer, published in 1976, it was concluded: a high degree of<br />

correlation between response and positive estrogen-receptor assay<br />

suggests the value of the diagnostic test as a means to select patients<br />

for tamoxifen treatment [7]. For the first time, we have a combination<br />

of a targeted drug and a predictive diagnostic assay/companion<br />

diagnostic test. Despite the fact that this study was published 35<br />

years ago, the conclusion about combining drug and diagnostics<br />

seems more relevant than ever.<br />

Late in the 1990s another example appeared, in fact the one which<br />

is most often referred in relation to personalised or stratified medicine,<br />

the combination of the IHC diagnostic test for HER2 and the<br />

monoclonal antibody trastuzumab. Trastuzumab targets the extracellular<br />

domain of HER2 and the effect is dependent on overexpression<br />

of the HER2 protein, or amplification of the HER2 gene, which<br />

is tested for by IHC or FISH, respectively [3, 8]. These protein or<br />

gene changes occur in 20–25% of all women with breast cancer,<br />

and it is in this subset of patients that trastuzumab has shown to be<br />

effective [9, 10]. Recently, trastuzumab has also proven to be effective<br />

in advanced gastric cancer, especially in the subgroup of<br />

patients that had the highest overexpression of the HER2 protein<br />

[11]. When it comes to HER2 positive breast cancer, a number of<br />

monoclonal antibodies and tyrosine kinase inhibitors have been<br />

developed or are under development, such as lapatinib, neratinib<br />

(Pfizer), and pertuzumab (Roche/Genentech), so the treatment of<br />

this patient group may be even more effective in the future [12, 13].<br />

Just as the development of trastuzumab must be regarded as a major<br />

breakthrough in the treatment of breast cancer, a new breakthrough<br />

seems to be under way in relation to the treatment of metastatic<br />

melanoma, where no effective treatment has so far been available.<br />

Very recently, promising interim phase III data with vemurafenib<br />

(Plexxicon/Roche) has been published [14]. Vemurafenib is a B-<br />

RAF kinase inhibitor that selectively blocks the RAF/MEK/ ERK<br />

pathway in BRAF V600E mutated cells [15]. It is estimated that<br />

approximately 40–60% of the melanoma patients have this type of<br />

BRAF mutation, which is measured by RT-PCR. In the phase III<br />

study vemurafenib was compared to dacarbazine in metastatic<br />

melanoma patients with the BRAF V600E mutation. The efficacy of<br />

vemurafenib was dramatically improved compared to dacarbazine,<br />

with a response rate of 48% for the patients treated with vemurafenib<br />

and only 5% for the dacarbazine group. The treatment with<br />

vemurafenib was further associated with a relative reduction of<br />

64% in the risk of death and a 74% reduction in the risk of disease<br />

progression compared with dacarbazine [14]. This data must be<br />

considered a major step forward in the treatment of this highly<br />

deadly disease and it will be interesting to see what the effect will<br />

be on the median overall survival when the study is finally concluded.<br />

Based on the recent phase III data, the FDA has (on 17 August<br />

<strong>2011</strong>) approved vemurafenib for the treatment of metastatic and<br />

unresectable melanomas together with the companion diagnostic<br />

test for BRAF V600E mutation.<br />

Conclusion<br />

If medical anticancer treatment is to be improved, disease heterogenecity<br />

must be taken into account when drugs are developed and<br />

subsequently used in the clinic. The drug-diagnostic co-development<br />

model has already proven its value with drugs like tamoxifen,<br />

trastuzumab, imatinib, and now also with vemurafenib. The use of<br />

companion diagnostics will be a decisive factor in utilising the clinical<br />

potential of the new targeted anticancer drugs to the benefit of<br />

patients.<br />

Authors<br />

Jan Trøst Jørgensen, MScPharm, PhD<br />

Dx-Rx Institute<br />

76 Baunevaenget<br />

DK-3480 Fredensborg, Denmark<br />

Maria Hersom, BSc<br />

Human Biology<br />

Faculty of Health Sciences<br />

University of Copenhagen<br />

DK-2200 Copenhagen, Denmark<br />

References<br />

1. Jørgensen JT. From blockbuster medicine to personalized medicine. Per<br />

Med. 2008;5:55-63.<br />

2. Collier R. Bye, bye blockbusters, hello niche busters. CMAJ.<br />

<strong>2011</strong>;183(11):E697-8.<br />

3. Jørgensen JT, Nielsen KV, Ejlertsen B. Pharmacodiagnostics and targeted<br />

therapies - a rational approach for individualizing medical anti-cancer therapy<br />

in breast cancer. Oncologist. 2007;12:397-405.<br />

4. Langreth R, Waldholz M. New era of personalized medicine - targeting<br />

drugs for each unique genetic profile. Oncologist. 1999;4:426-7.<br />

5. Jørgensen JT. Are we approaching the post blockbuster era?<br />

Pharmacodiagnostics and rational drug development. Expect Rev Mol<br />

Diagn. 2008;8(6):689-95.<br />

6. Cheung KL. The estrogen and progesterone receptors - setting the scene for<br />

pharmacodiagnostics. In: Jørgensen JT, Winther H, editors. Molecular diagnostics<br />

- the key driver of personalized cancer medicine. Singapore: Pan<br />

Stanford Publishing; 2010. p. 21-42.<br />

7. Lerner HJ, Band PR, Israel L, et al. Phase II study of tamoxifen: report of<br />

74 patients with stage IV breast cancer. Cancer Treat Rep.<br />

1976;60(10):1431-5.<br />

8. Jørgensen JT, Møller S, Rasmussen BB, et al. High concordance between<br />

two companion diagnostics tests: a concordance study between the<br />

HercepTest and the HER2 FISH pharmDx Kit. Am J Clin Pathol.<br />

<strong>2011</strong>;136: 145-51.<br />

9. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a<br />

monoclonal antibody against HER2 for metastatic breast cancer that overexpresses<br />

HER2. N Engl J Med. 2001 Mar;344(11):783-92.<br />

10. Gianni L, Dafni U, Gelber RD, et al. Treatment with trastuzumab for 1 year<br />

after adjuvant chemotherapy in patients with HER2-positive early breast<br />

cancer: a 4-year follow-up of a randomised controlled trial. Lancet Oncol.<br />

<strong>2011</strong> Mar;12(3):236-44.<br />

11. Jørgensen JT. Targeted HER2 treatment in advanced gastric cancer.<br />

<strong>Oncology</strong>. 2010;78(1):26-33.<br />

12. Roy V, Perez EA. Beyond trastuzumab: small molecule tyrosine kinase inhibitors<br />

in HER-2 positive breast cancer. Oncologist. 2009;14(11):1061-9.<br />

13. Lee-Hoeflich ST, Crocker L, Yao E, et al. A central role for HER3 in<br />

HER2-amplified breast cancer: implications for targeted therapy. Cancer<br />

Res. 2008 Jul;68(14):5878-87.<br />

14. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib<br />

in melanoma with BRAF V600E mutation. N Engl J Med. <strong>2011</strong><br />

Jun;364(26):2507-16.<br />

15. Bollaf G, Hirth P, Tsai J, et al. Clinical efficacy of a RAF inhibitor needs<br />

broad target blockade in BRAF-mutant melanoma. Nature. 2010<br />

Sep;467(7315):596-9.<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

13


Introduction<br />

<strong>Oncology</strong> is currently the fastest<br />

growing therapeutic area, and an<br />

increasing number of clinical<br />

trials are being conducted.<br />

Clinical trial management provides<br />

an excellent example of a<br />

multidisciplinary team approach.<br />

Indeed, the clinical research<br />

team will involve the sponsor,<br />

frequently its clinical research<br />

organisation (CRO) and devel-<br />

<strong>Cover</strong> <strong>Story</strong> - <strong>ESOP</strong>/<strong>NZW</strong> <strong>2011</strong> <strong>Congress</strong> <strong>Report</strong><br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

An assessment of the pharmacist’s role in<br />

oncology trial management<br />

This article reports on a <strong>ESOP</strong> survey which was undertaken to gather information about the current involvement<br />

of oncology pharmacists in clinical trial management across Europe. Significant differences in national<br />

practices are also highlighted and potential standardisation solutions are suggested.<br />

Mikael Daouphars<br />

PharmD, PhD<br />

opment specialists, the principal investigator and doctors, clinical<br />

research assistants and possibly research nurse(s), pharmacists,<br />

laboratory technicians and perhaps social workers.<br />

The pharmacist has become an integral part of the clinical<br />

research team. As a drug expert, he is the right person to<br />

manage all of the trial’s drug-related activities, thereby contributing<br />

to the overall smooth running of the study.<br />

Management of clinical trials in oncology<br />

By participating in clinical trials in oncology, the clinical pharmacist<br />

contributes to quality assurance. QuapoS 4 provides a<br />

reference guideline to be used to organise pharmacist-driven<br />

activities that are related to clinical trials [1]. On the basis of<br />

all applicable legal regulations, services that the pharmacist<br />

could provide in a clinical trial setting are:<br />

• investigational drugs ordering<br />

• shipment reception<br />

• storage<br />

• adequate labelling according to EU legislation if not previously<br />

realised by the sponsor<br />

• randomisation<br />

Ewelina Korczowska<br />

MPharm<br />

Martin Sevec<br />

PharmD<br />

• preparation of drugs, and eventual<br />

blinding<br />

• dispensing<br />

• return or disposal of used<br />

/unused investigational drugs.<br />

Pharmacists may also become<br />

involved in studies that investigate<br />

a drug’s stability, and the<br />

acceptance of patients who are<br />

selected to be enrolled on the clinical<br />

trial. Careful documentation<br />

of each of these steps in the clinical trial’s file is mandatory.<br />

European survey<br />

The role of pharmacy as a profession in the clinical research field<br />

needs to be fully understood. In recent years, European law has<br />

increasingly been implemented in national pharmaceutical law.<br />

However, practices can differ from one European country to<br />

another, as a result of differing legal regulations or historical pharmacy<br />

involvement. A study was therefore developed by <strong>ESOP</strong> to:<br />

• gather information about the current involvement of oncology<br />

pharmacists in clinical trial management in Europe<br />

• better clarify the level of pharmacy activities related to clinical<br />

trials in different European countries<br />

• identify appropriate actions to improve current standings.<br />

Methodology<br />

A short questionnaire was developed by three European pharmacists.<br />

All questions concerned pharmacist-related tasks<br />

during clinical trials, see Questionnaire. The questionnaire was<br />

circulated to all <strong>ESOP</strong> members with their online membership<br />

letter. It was also available on the <strong>ESOP</strong> website. Answers<br />

were collected from 21 December 2010 to 10 January <strong>2011</strong>.<br />

Figure 1: Profile of hospital respondents Figure 2: Number of oncology clinical trials<br />

University hospital<br />

General hospital<br />

<strong>Oncology</strong> hospital<br />

< 30<br />

30–50<br />

50–75<br />

75–100<br />

100–150<br />

> 150<br />

14 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


Questionnaire<br />

Tasks: medication/ancillaries order; cool chain medication<br />

transport verification; shipment delivery confirmation; temperature<br />

controlled medication storage; inventory review;<br />

medication preparation; billing; waste management including<br />

used medication containers destruction, expired medication<br />

destruction; communication with sponsor/CRO – monitoring<br />

visit, discrepancy reports, changes establishment; +/- subject<br />

randomisation; +/- medication reallocation.<br />

1. Name tasks different from the above mentioned that<br />

should/should not be your own. Does reality in your<br />

pharmacy overlap your opinion?<br />

2. Do you have independent ‘Department for clinical trial<br />

management’ or independent ‘Employee for clinical trial<br />

management’ in your pharmacy? If yes, what is its/his(her)<br />

scope (please specify in detail)?<br />

3. Do you have in your pharmacy transparent system of<br />

rewarding clinical trial staff according to its study workload?<br />

(In case you have independent ‘Department for clinical trial<br />

management’ or independent ‘Employee for clinical trial<br />

management’ in your pharmacy is it so all pharmacy<br />

employees are rewarded ‘equally’ taking into account their<br />

casual workload or those involved in clinical trial management<br />

are given an advantage over others?)<br />

4. Do you find better paper records or electronic ones (IVRS,<br />

IWRS)? Do you think they both have to be done or one is<br />

enough? Which one and why should then be preferred?<br />

5. Comparing clinical trial management with standard work<br />

do you think you need closer cooperation than just routine<br />

one with MDs (PI, subinvestigators) (please specify in<br />

detail)?<br />

6. When having a combination of orally medication and<br />

parenteral chemo in a single clinical trial do you in<br />

chemounits deal with orally medication as well or do you<br />

deal with parenteral chemo only and orally taken one is<br />

someone else responsibility (please specify in detail)?<br />

This questionnaire is the original version produced by the authors.<br />

PI: principal investigator.<br />

The completed questionnaires were returned by fax or were<br />

emailed as a PDF file.<br />

Results<br />

The results from each question have been analysed separately<br />

in order to highlight any differences or similarities between<br />

each European country. However, they have been presented<br />

holistically here for simplicity. Collected data can only be<br />

taken as an illustration of the current situation for the responding<br />

pharmacy. It cannot be taken as representative of the current<br />

situation for each European country in general.<br />

Thirty-seven completed questionnaires were received from 16<br />

European countries. Germany had the highest proportion of<br />

respondents (40% of the total). Approximately half of the<br />

respondents worked in general hospitals, one-third were from<br />

university hospitals, and only 8% were from specialist oncol-<br />

Table 1: Services provided by the pharmacy (% of<br />

respondents)<br />

100%<br />

Cool chain medication transport verification<br />

Shipment delivery confirmation<br />

Temperature controlled medication storage<br />

Medication preparation<br />

75–100%<br />

Inventory review<br />

Communication with sponsor/CRO<br />

Monitoring visit<br />

Medication order<br />

Waste management<br />

Expired medication destruction<br />

Expired/unused medication delivery to sponsor<br />

Discrepancy report<br />

50–75%<br />

Co-management of all ongoing hospital clinical trials<br />

Billing<br />

Relabelling<br />

Changes establishment<br />

25–50%<br />

Randomisation<br />

Medication reallocation<br />

Independent department for clinical trial management<br />

Transparent rewarding system<br />

ogy hospitals, see Figure 1. Most respondents (76%) had been<br />

involved with up to 50 clinical trials, see Figure 2.<br />

The pharmacy ward was involved in clinical trial management<br />

in 95% (n = 35) of the respondents’ hospitals. Pharmacy technicians<br />

were involved in approximately half of the hospitals.<br />

The mean number of pharmacists per hospital involved in each<br />

trial was between two and three, and between three and four<br />

for pharmacy technicians.<br />

Services provided by the pharmacy in relation to clinical trials<br />

are detailed in Table 1. There were few differences<br />

between countries, apart from relabelling, randomisation and<br />

medication reallocation. Billing was not systematic, and a<br />

transparent reward system was only reported by 25–50% of<br />

respondents. However, these relatively low figures do not<br />

mean that reward systems are generally absent. If the pharmacy<br />

staff are rewarded from the trial budget, the reward is<br />

shared equally without considering individual clinical trial<br />

management involvement.<br />

Electronic records of data were usually preferred by respondents<br />

over paper records, especially interactive internet web<br />

response services (IWRS). However, both paper and electronic—IVRS<br />

(interactive voice response systems)/IWRS—were<br />

often required by the sponsor.<br />

Discussion and conclusion<br />

Many oncology pharmacists currently operating in Europe<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

15


<strong>Cover</strong> <strong>Story</strong> - <strong>ESOP</strong>/<strong>NZW</strong> <strong>2011</strong> <strong>Congress</strong> <strong>Report</strong><br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

have extensive clinical trial management experience. This contribution<br />

has been widely recognised, with some tasks that have<br />

fallen to doctors now being allocated to the pharmacist.<br />

However, a transparent and equitable reward scheme for pharmacy-related<br />

tasks still needs to be standardised between countries.<br />

Communication between pharmacists and sponsors’ clinical<br />

research assistants could also be improved with regard to specific<br />

pharmacy-related tasks, especially the preparation of new<br />

drugs. The application of European regulations, along with the<br />

implementation of QuapoS recommendations and the collaboration<br />

of European pharmacists should help to standardise practice.<br />

Author for correspondence<br />

Mikael Daouphars, PharmD, PhD<br />

Pharmacy<br />

Cancer Centre Henri Becquerel<br />

Breast cancer radiotherapy is now<br />

established as a viable therapeutic<br />

strategy on a par with<br />

chemo- or hormone therapy.<br />

Principally, this is because lymph<br />

node-positive tumour irradiation not only<br />

reduces the frequency of tumour recurrence,<br />

but also improves overall survival. Modern<br />

irradiation technology is also now associated<br />

with fewer tolerability issues when compared<br />

with older radiotherapy techniques.<br />

There are also new recommendations regarding ductal carcinoma<br />

in situ (DCIS), specifically that radiotherapy is not necessary<br />

in the case of small malignant cells (< 2 cm), free resection<br />

margins (> 10 mm), and favourable gradings [1].<br />

Furthermore, radiotherapy with N+/- status has a positive<br />

effect on overall survival and recurrence reduction after both<br />

1 rue d’Amiens<br />

FR-76000 Rouen, France<br />

Co-authors<br />

Ewelina Korczowska, MPharm<br />

Przemienienie Panskie Clinical Hospital<br />

University of Medical Sciences<br />

Poznan, Poland<br />

Martin Sevec, PharmD<br />

University Hospital Motol<br />

Prague, Czech Republic<br />

Reference<br />

1. van Gemmern R. Quality standard for the oncology pharmacy services<br />

with commentary (QuapoS 4), 2008.<br />

Breast cancer irradiation: <strong>2011</strong> update<br />

Several developments have recently emerged in the field of breast cancer radiotherapy. This article summarises<br />

those that most impact pharmacists.<br />

Professor Dr Wolfgang Wagner<br />

MD, PhD<br />

mastectomy and breast-conservation surgery,<br />

see Figure 1 [2].<br />

To date, physicians have been cautious when<br />

indicating radiotherapy in patients with breast<br />

reconstructions immediately following mastectomy.<br />

This is because of a fear of implant failure.<br />

A recent study by Ho et al. has indicated<br />

that under certain circumstances, early radiotherapy<br />

may be possible despite implants—the<br />

trial showed a rate of removal of implants/<br />

replacements of 21% after seven years [3].<br />

The success of radiotherapy depends on the interval between<br />

surgery and the start of irradiation, a systematic review has<br />

suggested that this period should be a maximum of eight<br />

weeks, see Figure 2 [4]. Existing treatment programmes<br />

include primary chemotherapy, which can lead to a delay of<br />

Figure 1: Radiotherapy outcomes according to node status Figure 2:Associations between delay in postoperative radiotherapy<br />

for breast cancer and local recurrence rates<br />

16 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


Figure 3: Radiotherapy outcome versus tumour characteristics<br />

radiotherapy administration such that the risk of local recurrence<br />

is increased considerably, especially with regard to small<br />

tumour-free resection margins.<br />

Partially accelerated breast irradiation—total dose given within<br />

one week—is not yet considered standard therapy, whilst intraoperative<br />

radiotherapy is still regarded as being in an experiemental<br />

phase. Follow-up currently spans only two years, meaning that<br />

results cannot be conclusively evaluated, even though the<br />

observed local tumour control is comparable with the established<br />

procedure and toxicity appears to be lower.<br />

The results of radiotherapy depend on the tumour’s molecular<br />

characteristics cf. chemotherapy. In fact, when certain molecular<br />

characteristics are present, radiotherapy can be a disadvantage<br />

for the patient, see Figure 3 [5].<br />

Conclusion<br />

Breast cancer radiotherapy not only evokes local tumour con-<br />

Extravasation of cytotoxics: there is still room for improvement<br />

References (please see article on pages 18–19)<br />

1. Mader I, Furst-Weger P, Mader RM, Nogler-<br />

Semenitz E, Wassertheurer S. Extravasation of<br />

cytotoxic agents, 2nd edition. Wien New York:<br />

Springer; 2010.<br />

2. Parikh P, Pai VP, Ranjan S, Swami A,<br />

Cuchelkar V, Agarwal P, et al. Phlogenzym is<br />

safe and effective in reducing morbidity of<br />

vesicant chemotherapy extravasation - a<br />

prospective study. Int J Immunother. 2001;<br />

17(2-4):163-70.<br />

3. Langer SW, Sehested M, Jensen PB. Treatment<br />

Hospital workers perceptions about nano-technology<br />

References (please see article on pages 35–36)<br />

1. Siegrist M, Wiek A, Helland A, Kastenholz H.<br />

Risks and nanotechnology: the public is more<br />

concerned than experts and industry. Nat<br />

Nanotechnol. 2007 Feb 2 [cited <strong>2011</strong> Sep 28].<br />

Available from: www.nature. com/naturenanotechnology<br />

2. Toumey C. Rules of engagement. Nat Nanotechnol.<br />

2007 Jul 2 [cited <strong>2011</strong> Sep 28]. Available<br />

from: www.nature.com/naturenanotechnology<br />

of anthracycline extravasation with dexrazoxane.<br />

Clin Cancer Res. 2000;6:3680-6.<br />

4. Laurie SW, Wilson KL, Kernahan DA,<br />

Bauer BS, Vistnes LM. Intravenous extravasation<br />

injuries: the effectiveness of hyaluronidase<br />

in their treatment. Ann Plast Surg.<br />

1984;13:191-4.<br />

5. Mouridsen HT, Langer SW, Buter J, Eidtmann<br />

H, Rosti G, de Wit M, et al. Treatment of<br />

anthracycline extravasation with Savene (dexrazoxane):<br />

results from two prospective clinical<br />

3. Cobb M, Macoubrie J. Public perceptions about<br />

nanotechnology. Risks, benefits and trust. J<br />

Nanopart Res. 2004;6(4):395-405.<br />

4. Scheufele D, Lewenstein B. The public and nanotechnology:<br />

how citizens make sense of emerging<br />

technologies. J Nanopart Res. 2005;7:659-67.<br />

5. Currall SC. Nanotechnology and society new<br />

insights into public perceptions. Nat Nanotechnol.<br />

2009 Feb 4 [cited <strong>2011</strong> Sep 28]. Available from:<br />

www.nature.com/naturenanotechnology<br />

trol, but in certain circumstances, it can also extend overall<br />

survival. New irradiation technologies and programmes are<br />

likely to further improve results.<br />

Author<br />

Professor Dr Wolfgang Wagner, MD, PhD<br />

Professor of Radiotherapy<br />

Department of Radiotherapy<br />

Paracelsus Strahlenklinik Osnabrück<br />

69 Am Nantruper Holz<br />

DE-49076 Osnabrück, Germany<br />

References<br />

1. AGO Leitlinien 2010. AGO therapy guidelines ‘Breast Cancer’.<br />

Guidelines for diagnosis and treatment of breast cancer (Empfehlung<br />

seit 2002). Available from: www.ago-online.org<br />

2. McGale P, Darby S, Taylor C, Peto R. The 2006 worldwide overview<br />

of the effects of local treatments for early breast cancer on long-term<br />

outcome. Int J Radiat Oncol Biol Phys. 2006 Nov 1;66(3):S2-S3.<br />

3. Ho AY, Cordeiro P, Wright J, et al. Favorable long-term outcomes are<br />

achievable in women with immediate permanent implant breast reconstruction<br />

and postmastectomy radiation. Int J Radiat Oncol Biol Phys.<br />

2010 Nov 1;78(3):S3.<br />

4. Huang J, Barbera L, Brouwers M, Browman G, Mackillop WJ. Does<br />

delay in starting treatment affect the outcomes of radiotherapy? A systematic<br />

review. J Clin Oncol. 2003 Feb 1;21(3):555-63.<br />

5. Kyndi M, Sørensen FB, Knudsen H, Overgaard M, Nielsen HM,<br />

Overgaard J, et al. Estrogen receptor, progesterone receptor, HER-2,<br />

and response to postmastectomy radiotherapy in high-risk breast cancer:<br />

the Danish Breast Cancer Cooperative Group. J Clin Oncol. 2008<br />

Mar 20;26(9):1419-26.<br />

multicentre studies. Ann Oncol. 2007;<br />

18(3):546-50.<br />

6. Bertelli G, Gozza A, Forno GB, Vidili MG,<br />

Silvestro S, Venturini M, et al. Topical dimethylsulfoxide<br />

for the prevention of soft tissue<br />

injury after extravasation of vesicant cytotoxic<br />

drugs: a prospective clinical study. J Clin<br />

Oncol. 1995;13(11)2851-5.<br />

7. Alley E, Green R, Schuchter L. Cutaneous toxicities<br />

of cancer therapy. Curr Opin Oncol.<br />

2002;14:212-6.<br />

6. Miller CA, Cobb MD, Hays S. Public attitudes<br />

towards nanotechnology-enabled cognitive<br />

enhancement in the United States.<br />

7. Getting to know the public. Nat Nanotechnol.<br />

2009;4(2):71. [cited <strong>2011</strong> Sep 28]. Available<br />

from: www.nature.com/naturenanotechnology<br />

8. Kahan DM, Braman D, Slovic P, Gastil J, Cohen<br />

G. Cultural cognition of the risks and benefits of<br />

nanotechnology. Nat Nanotechnol. 2009;4(2):<br />

87-90.<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

17


<strong>Cover</strong> <strong>Story</strong> - <strong>ESOP</strong>/<strong>NZW</strong> <strong>2011</strong> <strong>Congress</strong> <strong>Report</strong><br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

Extravasation of cytotoxics: there is still room<br />

for improvement<br />

Introduction<br />

Extravasation of vesicant cytotoxics still remains a major challenge.<br />

In contrast to many expectations, the use of classic cytotoxic<br />

agents has actually increased in the ‘era’ of targeted therapies.<br />

Patients, nowadays, frequently experience fourth- and fifth-line<br />

treatments and enjoy longer survival and a decrease in cancer<br />

mortality rates. Thus, an individual patient receives many more<br />

IV infusions than two decades ago. This fact explains why<br />

extravasation is not a fading memory, but a reality that is here to<br />

stay in the years to come. To face extravasations, this article does<br />

not summarise the management procedures already outlined<br />

extensively in several excellent reviews and books [1], but instead<br />

proposes actions aimed at quality management and to discuss<br />

open scientific issues, which need to be urgently addressed. These<br />

suggestions include establishing an ‘Extravasation Task Force’ in<br />

your hospital to deal with the implementation of quality control<br />

and knowledge management.<br />

Risk factors for extravasation<br />

Several antineoplastic agents are associated with toxicity to cutaneous<br />

and subcutaneous tissue. The most common risk factors<br />

concern patient-associated and iatrogenic risk factors. With the<br />

hazard of irreversible complications after extravasation, prevention<br />

is of the utmost importance. A classification of antineoplastic<br />

agents addresses potential tissue damage: vesicant, irritant, and<br />

non-vesicant. Depending on the extravasated agent, a sequence of<br />

emergency measures need to be followed, which is best done by<br />

adhering to a standard operation procedure (SOP). There is good<br />

evidence for the successful use of antidotes for some antineoplastic<br />

agents. These antidotes are dimethylsulfoxide or<br />

hyaluronidase, often combined with topical measures such as<br />

cooling or application of heat, whereas the use of the anthracycline<br />

antidote dexrazoxane is still a matter of controversial discussion.<br />

Most relevant, novel compounds are poorly characterised<br />

regarding their toxic potential and are therefore to be considered<br />

as major drug-associated risks until clinical data are available.<br />

Building a task force in the institution<br />

There are several good arguments to concentrate expertise and<br />

action in a task force:<br />

• Splitting interventions between different departments and disciplines<br />

can result in contradictory measures and irreproducible<br />

management standards within one institution<br />

• Hardly comparable documentation<br />

• Expertise varies within departments that administer cytotoxics,<br />

Ursula Pluschnig, MD; Shahla Farokhnia, aHPh; Gunar Stemer, aHPh; Professor Robert M Mader, DSc<br />

As many patients receive more intravenous therapies than before, extravasation is a challenge that is here to<br />

stay for the foreseeable future. This article outlines several proposals for an improved quality management<br />

of these issues and discusses the science behind them.<br />

e.g. oncologic wards versus clinical units that occasionally<br />

handle infusions of cytotoxics.<br />

A possible approach is the formation of interdisciplinary clinical<br />

working groups with the following tasks:<br />

1. Members of the task force are the first port of call within the<br />

hospital<br />

2. Coordination of the subsequent treatment stages including the<br />

consultation of plastic surgeons<br />

3. In regular meetings (similar to a tumour board) the group<br />

members discuss individual cases of extravasation with the<br />

aim to define SOPs<br />

4. Training of hospital staff<br />

5. Dissemination of results by publication of novel data.<br />

Since the start of the task force at the Medical University of<br />

Vienna and the Vienna General Hospital in 2006, more than<br />

200 cases have been managed proving its usefulness and<br />

allowing the accumulation of the necessary expertise in a large<br />

hospital within very few years. According to our experience,<br />

the working group should include members from the disciplines<br />

of oncology, plastic surgery, oncology pharmacy, clinical<br />

pharmacology, pathology, nursing, and (as required) physiotherapy.<br />

Given the pharmacological expertise of hospital<br />

pharmacists working in oncology, they may provide valuable<br />

contributions to extravasation management, when physicians<br />

and nursing staff are in urgent need of support. It is a rewarding<br />

opportunity to further engage in interdisciplinary clinical<br />

activities for the patient’s benefit.<br />

Training staff members<br />

Regular training sessions help to raise awareness in the institution’s<br />

clinical members, medical, and nursing staff. Most<br />

importantly, they impart knowledge concerning a rapid, qualified,<br />

and appropriate intervention in a situation close to emergency.<br />

Documentation, documentation, documentation ...<br />

Standardised documentation is crucial to follow and evaluate the<br />

measures taken, possible sequelae and the final outcome. It is<br />

good practice to use one of the available documentation sheets<br />

accessible via the Internet, e.g. www.paravasate.at, offering forms<br />

in both English and German. Documentation in the minimal version<br />

includes description of the event, symptoms, measures taken,<br />

sequelae, and aftercare.<br />

18 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


Knowledge management<br />

Due to the lack of prospective clinical studies on extravasation of<br />

cytotoxic drugs, our current knowledge has to rely on individual<br />

case reports and animal studies. As this situation is not subject to<br />

change in the near future, creating evidence by adherence to SOP<br />

and stringent documentation is the hard way to overcome the current<br />

stagnation in this field.<br />

The science behind extravasation<br />

The type of damage associated with cytotoxic agents is a central<br />

issue, becoming prominent with every new, registered substance.<br />

Even the relatively simple classification into three types of damage<br />

is sometimes difficult, let alone to propose more sophisticated<br />

sub-classifications, e.g. addressing the fact that the vesicant<br />

potential of paclitaxel is not comparable to that of anthracyclines.<br />

After extravasation of vesicants, the clinical course and extent of<br />

damage can hardly be predicted. This is particularly true for anthracyclines,<br />

when necroses can develop even weeks after extravasation.<br />

Despite extensive pharmaceutical and pharmacological knowledge,<br />

there is no reliable method to predict tissue toxicity on the basis of<br />

physicochemical characteristics. Even when similar compound<br />

structures may be a helpful hint—as for the vinca alkaloids and the<br />

anthracyclines, clinical confirmation is always required.<br />

As extravasations are not always noticed immediately, a time delay<br />

longer than five days often elapses [2]. This immediately raises the<br />

question of the time period allowed before the efficiency of antidotes<br />

is compromised. Established antidotes have been tested for<br />

their use immediately after the extravasation event. Dexrazoxane<br />

was active against anthracyclines for at least three hours after the<br />

extravasation event, but activity was clearly reduced for daunorubicin<br />

after six hours [3]. Even shorter time windows seem to apply<br />

for the antidote hyaluronidase [4]. For this reason, instruction of<br />

patients and regular monitoring of IV infusions is of such crucial<br />

importance: early detection enables us to use instruments when<br />

time is the most critical factor for effectiveness.<br />

As dexrazoxane has been approved based on the data from two single-arm<br />

studies [5], it is not clear how its efficacy compares with that<br />

of the highly established clinical use of topical cooling dimethyl sulfoxide<br />

(DMSO), which has also been tested in a prospective clinical<br />

trial with a very similar rate of success [6]. This serious flaw in the<br />

study design makes it difficult to draw definite conclusions.<br />

Pathological changes within the affected tissue have been poorly<br />

characterised so far. Although many authors know that inflammatory<br />

processes are not the primary event after extravasation, it<br />

would be very useful to understand the histopathological changes<br />

in the skin in order to limit the use of corticosteroids, which are<br />

still frequently administered without real evidence.<br />

New therapeutic entities<br />

The pharmacological landscape has changed fundamentally over<br />

the last ten years and new oncology pharmacotherapy options will<br />

continue to emerge. With the introduction of mitosis-interfering<br />

compounds eribulin and vinflunin, two new chemical entities<br />

with novel mechanisms of actions, limited clinical experience and<br />

toxicological data enlarged the armamentarium of physicians. To<br />

date, few data regarding substance-specific risks and management<br />

procedures following extravasation exist. In the absence of conclusive<br />

data, the vesicant or irritant potential of new compounds<br />

can only be indirectly deduced from parameters such as structureactivity<br />

relationship and formulation characteristics (pH value,<br />

excipients, osmolarity of the infusion solution).<br />

At the same time, new galenic formulations of well-established<br />

compounds, e.g. liposomal daunorubicin and doxorubicin, and protein-bound<br />

paclitaxel are increasingly used due to decreased local<br />

and systemic toxicity compared to their non-modified precursors.<br />

Several monoclonal humanised antibodies, e.g. rituximab,<br />

trastuzumab, bevacizumab, cetuximab, panitumumab, and others,<br />

have become essential in the treatment of oncological and nononcological<br />

diseases. So far, clinical experience has shown type 1<br />

hypersensitivity reactions [7], which in rare cases result in severe<br />

side effects. Due to the mechanism of action, vesicant behaviour<br />

is highly unlikely, classifying them as low grade irritants.<br />

However, new humanised monoclonal antibodies, e.g. ipilimumab,<br />

brentuximab vedotin, emerge and for each individual compound<br />

the toxicity potential has to be evaluated again.<br />

Conclusion<br />

In summary, extravasation remains a dreaded complication of cytotoxic<br />

chemotherapy, at least for the next decade. Healthcare professionals<br />

engaged in extravasation management have to be aware of<br />

uncertainties and a possible lack of data regarding tissue toxicity of<br />

new compounds. This lack of knowledge should be addressed by an<br />

interdisciplinary ‘Extravasation Task Force’ engaging in the regular<br />

assessment of compounds, screening of literature, management of<br />

extravasation and their sequelae, adequate documentation, and publishing<br />

relevant information for the concerned scientific community.<br />

Authors<br />

Ursula Pluschnig, MD<br />

Professor Robert M Mader, DSc<br />

Clinical Division of <strong>Oncology</strong><br />

Department of Medicine I<br />

Medical University of Vienna<br />

18-20 Währinger Gürtel<br />

AT-1090 Vienna, Austria<br />

robert.mader@meduniwien.ac.at<br />

Shahla Farokhnia, aHPh<br />

Gunar Stemer, aHPh<br />

Pharmacy Department<br />

University Clinics<br />

Vienna General Hospital<br />

AT-1090 Vienna, Austria<br />

References<br />

References can be found on page 17.<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

19


Introduction<br />

DNA damage induced by endogenous and<br />

exogenous stimuli is a common event in<br />

every cell and its repair is routine because (as<br />

it is mandatory for cellular survival) efficient<br />

repair represents a strong survival advantage.<br />

The first report on DNA repair currently listed<br />

in MEDLINE dates from 1962 and<br />

describes the repair of UV-induced lesions.<br />

The knowledge that cells can recover from<br />

UV irradiation is even 30 years older than<br />

that. As there are a variety of different DNA<br />

lesions, cells have developed a matching variety of repair<br />

mechanisms some of which are interlinked and can serve as a<br />

backup for others if those happen to be ‘out of order’. Most of<br />

these mechanisms were discovered in the 1960s and 1970s [1].<br />

Classical DNA-targeting anticancer agents are just another<br />

source of DNA damage causing more lesions than usual during<br />

chemotherapy. Naturally, tumour and normal cells alike<br />

will attempt to repair the damage. Supposing that DNA repair<br />

enables cells to revert to their original state, repair in normal<br />

(stem) cells is a good thing because it reduces side effects,<br />

whereas in tumour cells it is unfavourable because it may help<br />

the cell to survive, which decreases the clinical response.<br />

However, there is increasing evidence that cells surviving<br />

DNA damage due to DNA repair do not necessarily maintain<br />

their genomic integrity, but instead sustain non-lethal mutations.<br />

If such a mutation conveys a growth advantage over<br />

other cells, it may give rise to a secondary tumour or to secondary<br />

resistance of the primary tumour.<br />

DNA repair pathways<br />

DNA damage affects the bases, or the ‘backbone’ of DNA.<br />

Damage to the bases is repaired either directly by O6-methylguanine-DNA-methyltransferase<br />

(MGMT), mismatch repair<br />

(MMR), base excision repair (BER) or nucleotide excision<br />

repair (NER). A damaged backbone is equivalent to a DNA<br />

single or double strand break and is repaired by homologous<br />

recombination (HR) or non-homologous end-joining (NHEJ).<br />

For a recent review, see Pallis et al. [2].<br />

The smallest alkyl residues, such as methyl and ethyl, are<br />

removed from the O6 position of guanine by MGMT which is<br />

also known as O6-alkylguanine DNA alkyltransferase (AGT).<br />

The modified guanine is directly reverted to its normal state,<br />

without the need to create and then fill a gap as with all other<br />

<strong>Oncology</strong> Pharmacy Practice<br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

DNA repair in cancer therapy: beneficial or<br />

harmful<br />

A lot of research is being performed on the modulation of DNA repair and its exploitation as a therapeutic principle<br />

to enhance the effects of chemotherapy. This paper outlines chemotherapy-induced DNA damage, its<br />

repair, consequences and the current state of DNA repair inhibitor development.<br />

Professor Dorothee C Dartsch<br />

PhD<br />

forms of DNA repair. Anticancer agents, such as<br />

temozolomide, pro- and dacarbazine, and<br />

nitrosoureas transfer small alkyl residues and may<br />

be more effective in the absence of MGMT.<br />

Bases modified by larger alkyl residues or oxidation<br />

are repaired by BER. The first step is the<br />

removal of the damaged base by a glycosylase,<br />

which leaves an intact backbone with an abasic<br />

site. Secondly, an apurinic or apyramidinic<br />

endonuclease cleaves the backbone and generates<br />

a strand break which is recognised by<br />

PARP. PARP enables binding of subsequent repair proteins<br />

such as polymerases and ligases. Synthesis of DNA along the<br />

intact strand is accomplished either by polymerase β‚ (can also<br />

be substituted by polymerase λ if only one base is replaced, or<br />

polymerases ε or δ that replace 2–13 bases. The overhanging<br />

flap of replaced DNA is removed by FEN1 (Rad27) before the<br />

XRCC1/ligase III complex joins the newly synthesised patch<br />

with the old strand.<br />

If the chemical modification is even bigger, e.g. a bulky adduct<br />

or a thymidine dimer, the damage is repaired by NER. In<br />

regions of the genome that are not actively transcribed, the heterodimer<br />

of xeroderma pigmentosum (XP) protein C and<br />

hHR23B is required for damage recognition (global genome<br />

repair). Where active transcription occurs, transcription-coupled<br />

repair is not necessary to induce the next steps which are<br />

similar in both regions. The transcription factor IIH, replication<br />

protein (RPA) and a set of XP proteins (A, G, B, D, G, and<br />

F) cooperate to separate the two DNA strands at the damaged<br />

site, to stabilise the opening, and to excise 27–29 bases around<br />

the modified one. Subsequently, like in BER, polymerases ε or<br />

δ fill the gap and ligase III and/or ligase I join the strands.<br />

Some forms of DNA damage, if unrepaired, can give rise to<br />

base mismatches and subsequent insertions and deletions during<br />

DNA replication. These are reverted by the MMR pathway.<br />

Recognition is accomplished by the protein MLH1 in varying<br />

heterodimeric combinations. The newly synthesised strand<br />

(identifiable by the lack of methylation) is incised and<br />

unwound by helicase. DNA exonuclease I excises the region<br />

around the mismatch, DNA polymerase δ fills the gap within<br />

the new strand and DNA ligase I seals the backbone.<br />

The DNA double strand break is considered the most hazardous<br />

lesion because just one of them can cause cell death if<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

21


it persists into mitosis, or genomic rearrangements if misrepaired.<br />

Two pathways have evolved that can accomplish<br />

repair of double strand breaks: HR and NHEJ. The pathway<br />

choice depends on different factors, e.g. cell type and cell<br />

cycle phase. In HR, recognition proteins named ATM and ATR<br />

recruit the ternary ‘MRN-complex’ that trims the 5’-ends at the<br />

break. This process is carried on by other enzymes and RPA<br />

binds to, and shelters, the generated single strand. In order to proceed<br />

with the repair, BRCA proteins 1 and 2 are needed to<br />

replace RPA with Rad 51. Binding of Rad 51 and/or XRCC 2 and<br />

3 proteins leads to alignment of the<br />

broken with a homologous intact<br />

strand. The latter serves as a template<br />

to re-synthesise the broken strand.<br />

HR also serves to repair interstrand<br />

crosslinks and lesions generated at<br />

stalled replication forks. NHEJ<br />

requires damage recognition by Ku<br />

70 and 80 proteins that bind to the<br />

ends of a break. They attract DNA-<br />

PKcs, an endonuclease that trims the<br />

ends of the broken strands and<br />

recruits polymerases λ and μ as well<br />

as DNA ligase IV that serve to fill the<br />

gaps and reseal the DNA backbones.<br />

An alternative form of NHEJ relies<br />

on PARP and the MRN complex to<br />

accomplish a limited resection of<br />

DNA ends at microhomology<br />

regions. Repair work is finished by<br />

the XRCC 1/ligase III complex. Thus, the alternative NHEJ pathway<br />

shares some key players with BER.<br />

Mutations in genes coding for DNA damage response-associated<br />

proteins are well known to cause severe disorders like<br />

ataxia telangiectasia, xeroderma pigmentosum, hereditary<br />

non-polyposis colorectal carcinoma, Bloom’s syndrome,<br />

BRCA-associated breast and ovarian cancers, and Fanconi<br />

anaemia.<br />

Chromosomal rearrangements and secondary<br />

malignancies<br />

Important knowledge about the long-term effects of anticancer<br />

chemotherapy comes from studies such as the British<br />

Childhood Cancer Survivor Study and the Childhood Cancer<br />

Survivor Study performed in the US and Canada. The first has<br />

followed, and is still following, a cohort of 17,981 five-year<br />

survivors of childhood cancer diagnosed with cancer at<br />

younger than 15 years between 1940 and 1991 in Great Britain<br />

(for the most recent publication see [3]). The second is a follow-up<br />

of 14,358 similar patients diagnosed between 1970 and<br />

1986 in the US and Canada (for the most recent publication see<br />

[4]). Of the childhood cancer survivors within these two studies,<br />

7.4% in the British and 9.6% in the American study had a<br />

diagnosis of a secondary neoplasm, which implies a cancer<br />

risk about four times higher than in the average, non-childhood<br />

<strong>Oncology</strong> Pharmacy Practice<br />

cancer population after a median follow-up time of 24.3 and<br />

23.0 years from diagnosis, respectively. The most common<br />

secondary tumour entities (in descending order) were central<br />

nervous system (CNS) tumours (mainly gliomas), non-malignant<br />

skin cancer, gastrointestinal, genito-urinary, breast, and<br />

bone cancers in the British study and non-malignant skin cancer,<br />

breast, thyroid, CNS cancer, sarcoma, bone cancer and<br />

leukaemia in the American study [5]. Moreover, data from<br />

those patients who survive the first have a very high risk of<br />

developing further, secondary neoplasms. Among the British<br />

cancer survivors with a secondary<br />

neoplasm, most had been treated for<br />

the primary tumour with alkylants or<br />

anthracyclines, some with epipodophyllotoxins<br />

or platinum agents, and<br />

sometimes as combined radiochemotherapy.<br />

The distribution of primary<br />

anticancer therapies was similar in the<br />

American study, as can be deduced<br />

from the freely accessible study data<br />

(ccss.stjude.org).<br />

Other studies have revealed that secondary<br />

malignancies are characterised<br />

by distinct genetic rearrangements.<br />

One example is the study from<br />

Aguilera et al. who reported that, of 22<br />

children with therapy-related myelodysplastic<br />

syndrome/acute myeloid<br />

leukaemia, none had normal cytogenetics<br />

[6]. The most frequent alteration was a del(7) genotype<br />

(all of these patients had received prior therapy with alkylating<br />

agents) and a t(9;11) genotype (all patients had received etoposide).<br />

On the other hand, patients treated with alkylating agents<br />

frequently show a set of unbalanced genetic aberrations: the loss<br />

of the whole chromosomes 5 and/or 7 (-5/-7 genotype) or the<br />

gain of a whole chromosome 8 (+8 genotype) [7]. Thus, one<br />

might suspect that mutagenic anticancer agents leave a ‘fingerprint’<br />

of typical genetic alterations.<br />

But how can there be characteristic translocations after the<br />

non-specific DNA damage inflicted by cytostatic compounds?<br />

To date, there are only vague hypotheses to explain<br />

this: the majority of random rearrangements may involve<br />

essential genes and, therefore, be lethal, which means that we<br />

do not see tumours with such rearrangements. Some agents<br />

have, if not a specific, then at least, a preferential site to<br />

cause damage, like the cleavage sites of topoisomerase II.<br />

The nuclear architecture may play a role in that it determines<br />

the proximity of certain chromosome bands and co-localisation<br />

with chromatin structural elements, e.g. topoisomerase<br />

II, scaffold attachment regions, regions of chromatin that are<br />

unprotected by histones and thus accessible for the transcription<br />

machinery and other enzymes working on DNA. Finally,<br />

an NHEJ signature has been found for all translocations<br />

known so far that consists of small deletions and duplications<br />

22 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


in each breakpoint, micro-homologies and non-template<br />

insertions [8].<br />

DNA repair inhibition as a therapeutic principle<br />

It is probably too simplistic to assume that DNA repair guarantees<br />

cellular survival, as we and others were able to show<br />

that (at least in vitro) cells still die despite DNA repair [9-11].<br />

Nevertheless, more and more studies suggest that it may be<br />

worthwhile to consider the effectiveness of the different repair<br />

pathways in order to guide the choice of the anticancer agent.<br />

For example, it has been shown that patients with a silent<br />

MGMT pathway benefit more from temozolomide, those with<br />

a low ERCC 1 level (NER pathway) from cisplatin, and those<br />

with an MMR defect from irinotecan, as opposed to patients<br />

with functioning repair pathways [12]. It is only a stone’s<br />

throw from this recognition to the idea that DNA repair might<br />

be systematically modulated in order to improve the effectiveness<br />

of chemotherapy.<br />

Inhibition of DNA repair, concomitant with a DNA-targeting<br />

chemotherapy is a compelling concept, not least because it<br />

may even possess some tumour specificity. Many tumours are<br />

characterised by abnormal DNA repair pathways. Thus, if<br />

there are two pathways A and B for a given DNA lesion and a<br />

tumour cell has a loss-of-function mutation for pathway A, it<br />

will be very vulnerable to DNA damage in the presence of an<br />

inhibitor of DNA repair pathway B, in contrast to a normal cell<br />

with two functioning repair pathways. This concept is called<br />

‘synthetic lethality’, and it is currently exploited mainly in<br />

BRCA 1/2 defective tumours. For a recent review see [13].<br />

This defect occurs frequently in hereditary forms of breast and<br />

ovarian carcinoma, but also in some sporadic tumours, such as<br />

triple negative (i.e. estrogen receptor (ER), progesterone receptor<br />

(PR) and HER2 negative) breast cancer. Cells with a BRCA<br />

1/2 defect are over-reliant on PARP to initiate repair at double<br />

strand breaks generated by stalled replication forks. Inhibition<br />

of PARP in these cells has been shown to augment the effect of<br />

anticancer agents. These findings have prompted researchers<br />

and companies to further develop DNA repair inhibitors.<br />

The group of DNA repair inhibitors that is furthest along the<br />

road towards the market are inhibitors of a repair enzyme<br />

called poly(ADP-ribose) polymerase, PARP. The three early<br />

ones are iniparib, olaparib, and veliparib, which are now in<br />

phase III clinical studies. The PARP inhibitors tested so far<br />

have shown fair tolerability alone and in combination with<br />

chemotherapy. However, initial euphoria about the new and<br />

promising target has been followed by some disappointment<br />

concerning iniparib because a phase III trial with more than<br />

500 patients with triple negative breast cancer did not match<br />

the hopes kindled by earlier studies [14]. This does not, however,<br />

eliminate the class of PARP and other DNA repair<br />

inhibitors from the list of promising, new biological response<br />

modifiers. For one thing, iniparib is probably the weakest of<br />

the inhibitors, for another, it may still be effective in specific<br />

subsets of patients with breast cancer and/or patients with<br />

other tumours that were not adequately represented in the<br />

study. Furthermore, PARP is not the only interesting target<br />

among the DNA repair proteins. Research is currently addressing<br />

other proteins like MGMT, the CHK1 and 2, ATM and<br />

ATR kinases, Rad 51, and probably many more.<br />

A very relevant question will be the long-term safety of treatment<br />

with any DNA repair inhibitor. Naturally, this is a completely<br />

black box at the moment, and it will take many years to<br />

find the answers, as we know from long-term studies with<br />

classical antineoplastic agents.<br />

Author<br />

Professor Dorothee C Dartsch, PhD<br />

Professor of Clinical Pharmacy<br />

Institute of Pharmacy<br />

University of Hamburg<br />

45 Bundesstrasse<br />

DE-20146 Hamburg, Germany<br />

References<br />

1. Ljungman DG, et al. The DNA damage response: repair or despair?<br />

Environ Mol Mutagen. 2010;51:879-89.<br />

2. Pallis AG, et al. DNA repair pathways and their implication in cancer<br />

treatment. Cancer Metastasis Rev. 2010;29:677-85.<br />

3. Reulen RC. Long-term risks of subsequent primary neoplasms among<br />

survivors of childhood cancer. JAMA. <strong>2011</strong>;305(22):2311-9.<br />

4. Armstrong GT, et al. Occurrence of multiple subsequent neoplasms in<br />

long-term survivors of childhood cancer: a report from the childhood<br />

cancer survivor study. J Clin Oncol. 29:3056-64.<br />

5. Meadows AT, et al. Second neoplasms in survivors of childhood cancer:<br />

findings from the childhood cancer survivor study cohort. J Clin<br />

Oncol. 2009;27:2356-62.<br />

6. Aguilera DG, et al. Pediatric therapy-related myelodysplastic syndrome/acute<br />

myeloid leukemia: the MD Anderson Cancer Center<br />

Experience. J Pediatr Hematol Oncol. 2009;31(11):803-11.<br />

7. Pedersen-Bjergaard, et al. Genetics of therapy-related myelodysplasia<br />

and acute myeloid leukemia. Leukemia. 2008;22:240-8.<br />

8. Schiavetti A, et al. Two secondary leukemias among 15 children<br />

given oral etoposide. Med Pediatr Oncol. 2001;37(2):148-9.<br />

9. Dartsch DC, et al. Repair of idarubicin-induced DNA damage: a<br />

cause of resistance? DNA Repair. 2007;6(11):1618-28.<br />

10. Schonn I, et al. Cellular responses to etoposide: cell death despite cell<br />

cycle arrest and repair of DNA damage. Apoptosis. 2010 Feb;15(2):<br />

162-72.<br />

11. Schonn I, et al. Ku70 and Rad51 vary in their importance for the<br />

repair of doxorubicin- versus etoposide-induced DNA damage.<br />

Apoptosis. <strong>2011</strong>;16:359-69.<br />

12. Martin SA, et al. Genomic instability and the selection of treatments<br />

for cancer. J Pathol. 2010; 220(2):281-9.<br />

13. Shaheen M, et al. Synthetic lethality: exploiting the addiction of cancer<br />

to DNA repair. Blood. <strong>2011</strong>:117(23):6074-82.<br />

14. Sanofi-aventis reports top-line results from phase III study with BSI-<br />

201 in metastatic triple-negative breast cancer [press release]. Sanofiaventis;<br />

<strong>2011</strong> Jan 27. [cited <strong>2011</strong> October 21]. Available from:<br />

http://en.sanofi.com/Images/13666_<strong>2011</strong>0127_BSI_en.pdf<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

23


Introduction<br />

Myelodysplastic syndromes (MDS) are a heterogeneous group of<br />

myeloid haematologic disorders characterised by aberrant, clonal<br />

haematopoietic stem cells producing abnormal red blood cells,<br />

platelets, and neutrophils. These abnormal cells never fully mature<br />

and lead to cytopenias with patient sequelae of severe anaemia as<br />

well as fatal bleeding and infections. There is also the risk of transformation<br />

to acute myeloid leukaemia in MDS. Myelodysplastic<br />

syndromes is a disease of the elderly. Thus, as people’s lifespan has<br />

increased, the incidence and prevalence of MDS has risen over the<br />

last several decades. In Europe and North America, the median age<br />

of MDS patients at presentation is 76 years. Moreover, the incidence<br />

of MDS increases with age with the annual incidence rising from 15<br />

to 50 cases per 100,000/year in those aged 70 years and older [1].<br />

Azacitidine (Vidaza®, Celgene Corporation, Summit, NJ, USA) is<br />

a cytidine nucleoside analogue and inhibitor of DNA methyltransferase<br />

[2-3], approved in the EU for treatment of higher-risk MDS,<br />

chronic myelomonocytic leukaemia (CMML, 10–29% marrow<br />

blasts without myeloproliferative disorder), and WHO-defined acute<br />

myeloid leukaemia with 20% to 30% blasts and multi-lineage dysplasia.<br />

Azacitidine has demonstrated the ability to prolong overall<br />

survival in patients with higher-risk MDS compared with conventional<br />

care [4].<br />

Azacitidine is rapidly degraded in water by hydrolysis [2-3]. Thus,<br />

per current EU Marketing Authorisation, azacitidine is to be reconstituted<br />

with sterile water for injection (WFI) to form a suspension<br />

and then administered within 45 minutes if stored at 25ºC. After<br />

reconstitution with sterile WFI, azacitidine suspension may be<br />

refrigerated (2–8ºC) for up to 8 hours. Reconstituted under these<br />

<strong>Oncology</strong> Pharmacy Practice<br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

®<br />

Cold water reconstitution of Vidaza ® with<br />

subsequent refrigerated storage prolongs<br />

drug stability<br />

Anthony Tutino1 , RPh; Mei Lai1,2 , PhD<br />

Abstract<br />

Study Objectives: This study assessed whether the stability of azacitidine suspension can be prolonged when reconstituted with<br />

refrigerated water for injection followed by storage under refrigerated conditions.<br />

Methods: Two lots of azacitidine (Vidaza) were reconstituted with refrigerated (2–8ºC) water for injection to form a suspension<br />

and immediately stored refrigerated (2–8ºC). After storage for 16, 18, 20, or 22 hours, azacitidine suspension was then placed at<br />

a constant 25ºC for 30 minutes then tested for potency, redispersibility time, and suspension appearance. Sterility testing was<br />

performed at the end of the study. Stability of azacitidine was defined as maintaining > 90% potency.<br />

Results: Azacitidine reconstituted with cold water (2–8ºC) followed by refrigerated storage (2–8ºC) and 30 minutes equilibration<br />

to 25ºC remained stable from baseline to 22 hours with a maximum loss of potency of 2.7% for each of the two lots of drug<br />

evaluated. At the 16, 18, 20, and 22 hour study time points and 30 minutes equilibration to 25ºC, redispersion time was 0.3 minutes<br />

with the appearance of fine white particles in suspension. All reconstituted vials stored refrigerated (2–8ºC) passed sterility<br />

testing at the end of study. Reconstitution of azacitidine with cold water for injection together with subsequent refrigerated storage<br />

is associated with a threefold prolongation in the stability time of the drug from 8 to 22 hours.<br />

Conclusion: This substantial increase in the time of azacitidine maintaining > 90% potency allows for prolonged in-use time that<br />

may provide for more convenience for pharmacists.<br />

Keywords: Azacitidine, cold water, reconstitution, stability, Vidaza<br />

conditions and times, azacitidine has been shown to remain stable (><br />

90% potency). However, recent evidence suggests that the drug<br />

degradation of azacitidine may be slowed with a decrease in temperature<br />

of the WFI used for reconstitution. The purpose of this study<br />

was to assess whether reconstituting azacitidine with refrigerated<br />

WFI (2–8ºC) followed by refrigerated storage at 2–8ºC would prolong<br />

the drug stability time of azacitidine.<br />

Analytical methods and stability testing<br />

The potency of azacitidine was determined by quantitating the sample<br />

against an external standard utilizing reverse phase chromatography<br />

with UV detection. This method has been fully validated in<br />

accordance with ICH Guidance Q2 (R1) [5]. The determination of<br />

suspension appearance for redispersibility time was performed visually,<br />

in accordance with ICH guidelines [6]. All of the analytical<br />

methods have been registered and accepted by the EMA for the testing<br />

of azacitidine.<br />

Stability testing of azacitidine was performed by utilising validated<br />

regulatory accepted analytical methods in accordance with ICH<br />

guidelines [7]. These studies have been conducted at conditions<br />

which encompass long-term, intermediate and accelerated storage of<br />

the drug product over a period of time. The results of these stability<br />

studies on azacitidine have demonstrated product stability and have<br />

been acknowledged by the EMA.<br />

Materials and methods<br />

Two lots of sterile commercial scale azacitidine drug product,<br />

each nearing the end of their expiry period of 48 months, were<br />

used for the study. Each lot of drug product was prepared as a<br />

homogeneous mixture of lyophilized powder and placed in<br />

24 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


Table 1: Averaged* Azacitidine assay† Stability‡ after cold WFI reconstitution and refrigerated storage (2–8°C) from<br />

baseline (0 hours) to 22 hours<br />

Lot 1<br />

Baseline Average % 16 hours Average % 18 hours Average % 20 hours Average % 22 hours Average %<br />

for 3 vials* assay at BL for 3 vials* assay at 16 for 3 assay at for 3 assay at for 3 assay at<br />

hours vials* 18 hours vials* 20 hours vials* 22 hours<br />

Assay 95.3% 95.5% 93.7% 93.9% 94.2% 93.6% 91.6% 92.5% 92.5% 92.8%<br />

94.6% 94.3% 93.2% 93.0% 92.9%<br />

96.7% 93.7% 93.6% 92.9% 93.0%<br />

% loss of – 1.6% 1.9% 3.0% 2.7%<br />

assay from<br />

BL<br />

Lot 2<br />

Assay 95.2% 95.2% 94.6% 93.7% 93.6% 92.7% 92.8% 92.9% 93.7% 92.5%<br />

94.5% 93.2% 93.3% 92.6% 91.9%<br />

95.9% 93.3% 91.1% 93.4% 91.9%<br />

% loss of – 1.5% 2.5% 2.3% 2.7%<br />

assay from<br />

BL<br />

*Each averaged per cent comes from 3 vials of product; †% per EU Marketing Authorisation; ‡Stability of Vidaza based on maintenance of > 90% potency.<br />

WFI, water for injection.<br />

labelled vials in preparation for reconstitution. Vials from each<br />

lot of azacitidine drug product were then reconstituted with 4<br />

mL of refrigerated (2–8ºC) WFI and immediately placed in<br />

refrigerated storage (2–8ºC). After refrigerated storage for 16, 18,<br />

20, and 22 hours, reconstituted vials of azacitidine were removed<br />

from refrigeration and placed at 25ºC for 30 minutes.<br />

Equilibration time to 25ºC for refrigerated azacitidine suspension<br />

has been previously determined to be 30 minutes per EU<br />

Marketing Authorisation. After 16, 18, 20, and 22 hours of refrigerated<br />

storage and 30 minutes equilibration to 25ºC, the reconstituted<br />

vials were shaken vigorously and tested for potency,<br />

redispersibility time, and appearance of the suspension. Stability<br />

of azacitidine was defined as maintaining > 90% potency.<br />

Sterility of the refrigerated drug product was assessed for reconstituted<br />

vials stored at 2–8ºC at end of study.<br />

Results<br />

After cold water (2–8ºC) reconstitution and refrigerated storage<br />

(2–8ºC) followed by 30 minutes equilibration to 25ºC, azacitidine<br />

remained stable from baseline to 22 hours with a maximum loss of<br />

potency of 2.7% for each of the 2 lots of drug product evaluated, see<br />

Table 1. At the 16, 18, 20, and 22 hour study time points and 30<br />

minutes equilibration to 25ºC, redispersion time was 0.3 minutes<br />

with the appearance of fine white particles in suspension and the<br />

absence of clumps. All reconstituted vials stored refrigerated (2–8ºC)<br />

passed sterility testing at end of study.<br />

Discussion<br />

Because azacitidine readily degrades in water after reconstitution,<br />

the drug—per the EU Marketing Authorisation—must be administered<br />

within 45 minutes when stored at 25ºC and within 8 hours<br />

under refrigerated conditions to assure maintenance of potency of<br />

> 90%. The results of this trial, however, show that the stability<br />

(> 90% potency) and usage time of reconstituted azacitidine can be<br />

prolonged nearly threefold from 8 hours to 22 hours using cold water<br />

(2–8ºC) reconstitution followed by refrigerated (2–8ºC) storage.<br />

This reconstitution procedure showed no effects on redispersibility,<br />

appearance, or sterility after the prolonged refrigerated storage time.<br />

These findings have important implications for providing expanded<br />

usage time with azacitidine for pharmacists and other caregivers.<br />

Conclusion<br />

Reconstitution using cold WFI followed by refrigerated storage is<br />

associated with a threefold prolongation in the stability time of azacitidine<br />

(8 to 22 hours). This substantial increase in the time of azacitidine<br />

maintaining > 90% potency allows for prolonged in-use time<br />

that may provide for more convenience for pharmacists.<br />

Acknowledgements<br />

The authors wish to acknowledge the assistance of Acceleration<br />

Laboratory Services, Inc, 2634 NE Hagan Road, Lee’s Summit,<br />

MO 64064, USA, in the conduct of this study and the writing assistance<br />

of Mr Neil Malone, MA, of Celgene Corporation, in the development<br />

of this manuscript.<br />

Author for correspondence<br />

Anthony Tutino 1 , RPh<br />

1 Celgene Corporation<br />

Pharmaceutical Development<br />

110 Allen Rd<br />

Basking Ridge, NJ 07938, USA<br />

Tel: +1 908 8604071<br />

Fax: +1 908 8604079<br />

atutino@celgene.com<br />

Co-author<br />

Mei Lai 1,2 , PhD<br />

2 Clovis <strong>Oncology</strong>, Pharmaceutical<br />

Development, Boulder, CO, USA<br />

References<br />

References can be found on page 34.<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

25


<strong>Oncology</strong> Pharmacy Practice<br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

A practical approach to oral tumour therapy<br />

Oral anti-tumour drugs present many challenges for medical staff, pharmacists, and patients [1]. A recent German<br />

Society for Oncological Pharmacy (DGOP) initiative sought to address some of the more urgent questions in this<br />

complex field [2]. This article summarises the collaborative thoughts of a haemato-oncologist and a pharmacist.<br />

The problem<br />

Oral anti-tumour drugs are commonly considered<br />

to be less aggressive, less toxic, and less<br />

problematic than their IV counterparts.<br />

However, despite the long-term availability of<br />

some of these agents, there is no recent guidance<br />

regarding the administration of these drugs. In<br />

order to achieve optimal patient outcomes, it is<br />

vital that information is constantly updated. This<br />

helps to ensure that well established compounds<br />

such as busulfan or cyclophosphamid, and fairly<br />

new targeted therapies such as imatinib and everolimus, continue<br />

to be used optimally. Some of the most commonly available oral<br />

tumour therapies are listed in Table 1.<br />

This article focuses on small (low) molecular (weight) kinase<br />

inhibitors (SMKIs), considered by many experts to be the ‘rising<br />

stars’of personalised targeted therapy. SMKIs are considered to be<br />

highly selective against tumour cells, but they can also have<br />

severe, sometimes life-threatening side effects. Their toxicities<br />

affect most organs, and include carcinogenic, mutagenic, and toxic<br />

to reproduction effects. These toxicities result from inhibiting not<br />

only tumour-dependent kinases in the tumour cells, but also the<br />

normal variant counterparts in healthy cells, exactly as classical<br />

chemotherapy affects both the tumour and host organism.<br />

Compliance<br />

Oral (chemo-) therapy relies heavily on patient compliance.<br />

Physicians and nurses must therefore work together to explain the<br />

therapy to the patient, and counter any individual fears and side<br />

Jürgen Barth<br />

Table 1: Classes and INN names of oral anti-tumour therapeutics<br />

Alkylating agents Antimetabolites Podophyllotoxine IMIDs SMKI<br />

derivatives<br />

Busulfan Capecitabine Etoposide Lenalidomide Dasatinib<br />

Chlorambucil Fludarabinphosphate Topoisomerase I Pomalidomide* Erlotinib<br />

inhibitor<br />

Cyclophosphamide Methotrexate Topotecan Thalidomide Everolimus<br />

Estramustine Mercaptopurine Topoisomerase II Gefitinib<br />

inhibitor<br />

Lomustine S 1 Idarubicin Imatinib<br />

Melphalan Tegafur-uracil Vinca-alkaloides Lapatinib<br />

Procarbazine Tioguanine Vinorelbine Nilotinib<br />

Temozolomide Other Pazopanib<br />

Treosulfan Hydroxycarbamid Sorafenib<br />

Trofosfamide Mitotane Sunitinib<br />

Hormone/anti- Vandetanib<br />

hormones<br />

IMIDs: immune modulatory drugs; *Pomalidomide not yet approved.<br />

effects. Oral therapy may lead to an increased<br />

quality of life and greater self-management, but if<br />

this type of therapy is mismanaged, it can evoke<br />

serious side effects and become burdensome.<br />

Food<br />

The bioavailabilities of oral cytostatics vary extensively,<br />

depending on the timing of concomitant<br />

food intake. The effect of fat or other food components<br />

may result in over or underdosing, depending<br />

on the exact drug administered. Table 2 provides<br />

an overview of recommended oral SMKI administrative timing<br />

in relation to food intake.<br />

Knowledge<br />

Patients need to know the advantages and disadvantages of<br />

their therapy, their exact dosing requirements, and why they<br />

should not discontinue therapy. To achieve these objectives,<br />

doctors and pharmacists must work together, the therapy must<br />

be practical enough to fit in with the patient’s daily routine,<br />

and the patient must be informed of the exact circumstances<br />

when he/she should contact his/her physician. During the<br />

patient consultation, however time-consuming or demanding it<br />

may turn out to be, it is essential to ensure that the patient<br />

agrees to the therapy and that the information provided is not<br />

intimidating, overwhelming, or complicated.<br />

Interactions<br />

Concomitant intake of medicines with food and/or other drugs<br />

may result in unwanted interactions between the cytochrome P450<br />

superfamily of metabolic enzymes.<br />

However, these interactions<br />

only become important if they<br />

result in a clinically significant<br />

effect, e.g. an inhibition of degradation<br />

that causes a 2- to 5-fold<br />

difference in predicted plasma<br />

levels. Like approximately 50%<br />

of therapeutic entities, SMKIs are<br />

metabolised by CYP3A4. When<br />

two molecules are simultaneously<br />

metabolised by CYP3A4, it is<br />

difficult to predict which one will<br />

take precedence. As a practical<br />

approach, all medicines and supportive<br />

agents such as vitamin<br />

or mineral tablets must be<br />

reported, the patient must be<br />

informed of any potential inter-<br />

26 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


actions, and if applicable he/she must refrain from their concomitant<br />

medication.<br />

Table 2: Recommended SMKI administration in relation<br />

to food intake<br />

To be taken with<br />

food<br />

To be taken<br />

without food<br />

No matter<br />

Imatinib Erlotinib<br />

Everolimus<br />

Lapatinib<br />

Nilotinib<br />

Pazopanib<br />

Sorafenib<br />

Dasatinib<br />

Gefitinib<br />

Sunitinib<br />

Vandetanib<br />

Side effects and toxicity management<br />

SMKIs that target the epidermal growth factor receptor<br />

(EGFR) and the anti-EGFR-monoclonal antibodies, also act<br />

against the EGFR in normal skin. This can evoke dermatotoxicities<br />

such as inflammation and acne-like efflorescence, without<br />

the bacterial infection. Firstly, the skin changes in a way<br />

similar to acne, then it becomes dry, and finally it is rendered<br />

very sensitive and dry. There are many recommendations<br />

available for reversing all of these three phases [3-5].<br />

There is some evidence to suggest that RAF-kinase inhibitors<br />

may evoke secondary malignancies such as carcinoma of the<br />

epithelium [6]. As a consequence, pharmacists, physicians,<br />

and nurses should monitor closely any changes in the patient’s<br />

skin.<br />

SMKI inhibition can also adversely affect several healthy<br />

kinase variants in the heart [7]. The fusion protein BCR/Abl is<br />

the molecular driver of chronic myeloid leukaemia, and<br />

because dasatinib, imatinib and nilotinib not only inhibit<br />

BCR/Abl but also the unmutated form of the Abelson-kinase<br />

that is essential for cardiomyocyte survival, heart function can<br />

become diminished. Off-target cardiotoxicity has also been<br />

reported with other SMKIs, including RSK, RAF, and HER2.<br />

As a consequence, heart function should be monitored for the<br />

duration of SMKI therapy. This includes the monitoring of QT<br />

changes, a sign of arrhythmia.<br />

Other organs that can be affected by SMKI toxicity are the thyroid<br />

gland (sunitinib, pazopanib) and the liver [8]. Blood pressure is<br />

influenced by all anti-angiogenic agents, e.g. vascular endothelial<br />

growth factor inhibitors, and routine blood pressure monitoring is<br />

therefore necessary. Class effects include hypertension and venous<br />

thrombosis.<br />

Gastrointestinal toxicity<br />

All known ATP mimetics cause (sometimes dose-limiting) diarrhoea,<br />

and some degree of nausea and vomiting. Many patients<br />

seem uneasy talking about diarrhoea. Initially, patients need to<br />

understand the definition of diarrhoea. Grades of diarrhoea are<br />

clearly defined in the Common Terminology Criteria for<br />

Adverse Events version 4.0, 28 May 2009, by the number of<br />

stools per day, and if applicable, ostomy output. Intervention<br />

can include the use of loperamide. Ciprofloxazin use is mandatory<br />

if diarrhoea persists for more than 48 hours. Octreotid can be<br />

regarded as rescue medication if symptoms continue to persist.<br />

Conclusions<br />

• Close collaboration between the physician, nurse and pharmacist<br />

is essential for guiding the patient through oral chemotherapy.<br />

• Medical and pharmaceutical staff should be highly educated<br />

about oral anti-tumour therapies.<br />

• Prescriptions should always be dispensed on time.<br />

• Physicians and pharmacists should share patient information and<br />

ensure that it is as helpful as possible.<br />

• Oral tumour therapy is complex, and all healthcare stakeholders<br />

should work together.<br />

• Confidence and clarity are mandatory when advising patients<br />

about the importance of compliance.<br />

• All recommended supportive medications should be closely scrutinised,<br />

and should be chosen after careful consideration of the<br />

patient’s individual situation—an inability to do so may be counter-productive.<br />

Author<br />

Jürgen Barth<br />

Specialist in Clinical Pharmacy and <strong>Oncology</strong> Pharmacy<br />

Justus Liebig University<br />

Medizinische Klinik IV<br />

Universitätsklinik<br />

36 Klinikstrasse<br />

DE-35385 Giessen, Germany<br />

References<br />

1. Barth J. Kompetente Antworten auf scheinbar leichte Fragen. Onkologische<br />

Pharmazie. 2009;11(3):4-7.<br />

2. Deutsche Gesellschaft für Onkologische Pharmazie (DGOP). Orale<br />

Zytostatikatherapie — sicher und effektiv durch gemeinsame Beratung.<br />

Available from: www.dgop.org/anmeldung_kampagne.php<br />

3. Lacouture ME. Mechanisms of cutaneous toxicities to EGFR inhibitors.<br />

Nat Rev Cancer. 2006;(10):803-12.<br />

4. Potthoff KM, Hassel JC, Wollenberg A, et al. Therapie und Prophylaxe<br />

EGFR-Inhibitor-induzierter Hautreaktionen. Arzneimitteltherapie. 2010;<br />

28:191-8.<br />

5. Schimanski CC, et al. Cetuximab-induced skin exanthema: Improvement<br />

by a reactive skin therapy. Mol Med <strong>Report</strong>. 2010;3(5):789-93.<br />

6. Dubauskas Z, et al. Cutaneous squamous cell carcinoma and inflammation<br />

of actinic keratoses associated with sorafenib. Clin Genitourin Cancer.<br />

2009;7(1):20-3.<br />

7. Force T, Krause DS, Van Etten RA. Molecular mechanisms of cardiotoxicity<br />

of tyrosine kinase inhibition. Nat Rev Cancer. 2007;7(5):332-44.<br />

8. Loriot Y, et al. Drug insight: gastrointestinal and hepatic adverse effects of<br />

molecular-targeted agents in cancer therapy. Nat Clin Pract Oncol.<br />

2008;5(5):268-78.<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

27


<strong>Oncology</strong> Pharmacy Practice<br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

Setting up a pharmacist-led chemotherapy<br />

clinic: a practical guide<br />

In the UK, several pharmacist-led chemotherapy clinics have been reported. The increasing use of oral<br />

chemotherapy is revolutionising the way that chemotherapy services are provided. This article is a practical<br />

guide for oncology pharmacists who are considering setting up a chemotherapy clinic.<br />

Introduction<br />

In the UK, pharmacists and nurses have been<br />

able to train as prescribers since 2003.<br />

Initially, this was in partnership with a doctor<br />

(called supplementary prescribing), but more<br />

recently, pharmacists have been able to prescribe<br />

independently. Under current legislation,<br />

trained pharmacists can prescribe the<br />

full range of licensed and unlicensed medicines,<br />

with the exception of controlled drugs<br />

such as opiates.<br />

The chemotherapy clinic at Airedale Hospital, West Yorkshire,<br />

UK, has been running since before these changes were introduced—a<br />

doctor was required to sign all the prescriptions<br />

which were prepared by the pharmacist. The introduction of<br />

pharmacist prescribing simplified the pathway and allowed<br />

development of new services.<br />

In the UK, several important national documents have highlighted<br />

the risks of chemotherapy treatment and advised services<br />

to look for innovative ways of working to ensure safer<br />

treatment for patients [1, 2]. The introduction of pharmacistled<br />

chemotherapy clinics is one way in which some services<br />

are doing this [3-5].<br />

In addition, the introduction of consultant oncology pharmacists<br />

means that pharmacists are well placed to take on some<br />

of the roles more traditionally associated with medical oncologists<br />

[6, 7].<br />

This article is a brief practical guide for oncology pharmacists<br />

who are considering setting up a chemotherapy clinic.<br />

The groundwork<br />

The first consideration should always be to think about<br />

whether a pharmacist-led clinic is required within the local<br />

service. There must be clear benefits for patients and for the<br />

service. It is vital that discussions take place between pharmacists,<br />

oncologists, nurses and managers. Some benefits of<br />

pharmacist-led clinics are shown in Table 1.<br />

It is useful to include patients in discussions about new services;<br />

they can give useful insights into what they want as users<br />

of the new service. When clinical and managerial support has<br />

been established, a robust business case is required. This<br />

details the costs involved in setting up the service to include<br />

Carl Booth, BPharm, MPhil,<br />

MRPharmS IP<br />

staff costs and facilities and also any income<br />

generated by the clinic. Think about what backfill<br />

may be required to cover the pharmacist’s<br />

other duties and who will cover the service in<br />

the event of holiday or sickness.<br />

If there are any similar clinics already running<br />

within the local area, arrange to visit them and<br />

speak to the staff who have already been<br />

through the process. Colleagues are usually<br />

willing to give you advice on where things went<br />

well or badly.<br />

A very important point is to audit the current service before<br />

you make any changes. This will give you some useful information<br />

to demonstrate the benefits of your new service at a<br />

later date.<br />

Setting up the new clinic<br />

It is important to identify the group of patients you will see in<br />

the clinic. In large centres this might be a specific group, such<br />

as colorectal cancer patients on capecitabine. In smaller centres<br />

it might be all capecitabine patients, or patients on oral<br />

chemotherapy treatment. Remember that you may not see<br />

many patients at first, but if the clinic is a success, the numbers<br />

will build up over time and you will need to plan ahead so that<br />

you are not over booking clinics, leading to delays for patients<br />

and stress for staff!<br />

Consider which staff need to be involved in the clinic. Will<br />

you be running it alone or in partnership with an oncology<br />

nurse or medical oncologist, and how will patients be referred<br />

to your clinic?<br />

Think about the facilities that will be required. There may be<br />

space in the outpatient clinic or on the chemotherapy day unit.<br />

Table 1: Benefits of pharmacist-led chemotherapy clinics<br />

Patient benefits:<br />

• direct access to a medicines expert<br />

• quicker access to medicines<br />

• shorter waiting times<br />

Service benefits:<br />

• reduced costs compared with medical oncology clinics<br />

• increased capacity of medical clinics<br />

• reduced workload on chemotherapy day units<br />

• improved team working and job satisfaction<br />

• compliance with national standards<br />

28 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


Ensure that you have access to IT services such as blood<br />

results or prescribing systems.<br />

It is essential that protocols are in place before you start to<br />

see patients in the clinics. This will ensure that you work<br />

within locally agreed boundaries, ensure the safety of<br />

patients and support the staff. You will need access to the<br />

patient record, which may be paper or electronic medical<br />

notes. In our chemotherapy consent clinic we have individual<br />

patient plans for each regimen and disease state. The<br />

medical oncologist signs the correct plan when the decision<br />

to start chemotherapy is made in the outpatient clinic and<br />

this acts as a referral to the pharmacy and nurse-led<br />

chemotherapy consent clinic and also as a treatment plan for<br />

prescribing chemotherapy.<br />

Check that you have authorisation to undertake clinical tests<br />

that you might need to order. If not, then request permission<br />

from your employing hospital or put systems in place to<br />

ensure that these tests are ordered in advance. In some cases<br />

you may require test results to be available on the day of the<br />

clinic in order to prescribe chemotherapy. It may be useful<br />

to get these done the day before to ensure that the patient is<br />

not waiting.<br />

Running the clinic<br />

Once the clinic details have been finalised and the clinic is<br />

ready to start, make sure that patients are clear about whom<br />

they will be seeing in the clinic. If patients are expecting to see<br />

their medical oncologist they may be concerned to see a pharmacist<br />

in the clinic. Agree the wording of clinic letters with the<br />

medical secretaries to make sure the location and timing of the<br />

clinics is clear. If you want patients to bring their medicines, or<br />

a list of medicines, then this request should be added to the<br />

clinic letter.<br />

An important point to consider is how you will access medical<br />

assistance if needed. If you see a patient in the clinic who<br />

becomes acutely unwell or who exhibits signs of illness that<br />

require medical assessment, how will this be arranged? It may<br />

be an informal arrangement with the medical oncologist or oncall<br />

physician to call if needed, or it may be that you run your<br />

clinic alongside an oncologist so that assistance is always on<br />

hand.<br />

Make sure that all your actions with the patient are documented<br />

in the medical record. You may need to be able to justify<br />

your actions at a later date.<br />

Reviewing the service<br />

Once you have your clinic up and running, it is not time to sit<br />

back and relax! Consider the first few clinics as a pilot, following<br />

which you may wish to make changes in how the clinic<br />

runs. Think about what went smoothly and what could have<br />

been done better. Did the clinics run to time and did you have<br />

access to everything you needed?<br />

Once the clinic is established as a long-term service then consider<br />

auditing to establish whether the proposed benefits to<br />

patients and to the service have been achieved. Compare your<br />

results with the baseline audit which you carried out before<br />

setting up the service. Consider undertaking a patient satisfaction<br />

survey.<br />

Outcomes should be presented locally, for example at clinical<br />

governance meetings. It is important to disseminate your<br />

results more widely at meetings and conferences. Positive outcomes<br />

from your new clinic will assist other pharmacists in<br />

making the case for their new clinic.<br />

Hopefully your new clinic will become a beacon of clinical<br />

oncology pharmacy practice. You can use it to train junior<br />

pharmacists and senior colleagues alike. Where patients have<br />

a choice about where they are treated it may also be used to<br />

attract patients to your oncology service. Finally, it may be<br />

used to attract and retain oncology pharmacists where they can<br />

use the clinic to develop their knowledge and skills in oncology<br />

working towards advanced levels of clinical practice.<br />

Author<br />

Carl Booth, BPharm, MPhil, MRPharmS IP<br />

Lead Pharmacist<br />

Cancer Services and Clinical Trials<br />

Airedale NHS Foundation Trust<br />

Pharmacy Department<br />

Airedale General Hospital<br />

Skipton Road, Steeton, Keighley<br />

West Yorkshire BD20 6TD, UK<br />

References<br />

1. Department of Health. Systemic anti-cancer chemotherapy, for better<br />

or worse? A report by the National Confidential Enquiry into Patient<br />

Outcome and Death. London: DH; 2008.<br />

2. Department of Health. Chemotherapy services in England: ensuring<br />

quality and safety. A report from the National Chemotherapy Advisory<br />

Group. London: DH; 2009.<br />

3. Booth CD, Dyminski P, Jeyasangar G. Chemotherapy Consent Clinic.<br />

Healthcare Pharmacy. Apr 2005;2(2):24-5. Available from: www.nelm.<br />

nhs.uk/en/NeLM-Area/Evidence/Medicines-Management/References/<br />

2005 May/24/Chemotherapy-consent-clinic/<br />

4. Donovan G, Evans S. Development of nurse/pharmacist-led clinic for<br />

the treatment of breast cancer. Adv Breast Cancer. 2005; November:<br />

47-9.<br />

5. Bowden EM, Horne N. 59 A joint non-medical prescribing clinic for<br />

the management of patients requiring tyrosine kinase inhibitors and<br />

chemotherapy treatments led by a lung cancer clinical nurse specialist<br />

and a Macmillan pharmacist. Lung Cancer. 71 (Suppl. 1), <strong>2011</strong>, S20.<br />

doi:10.1016/S0169-5002(11)70059-9<br />

6. Department of Health. Guidance for the development of consultant<br />

pharmacist posts. London: DH, 2005.<br />

7. Kirk S. Diversity of a consultant oncology pharmacist’s role. British<br />

Journal of Clinical Pharmacy. 2010;(2):23-4.<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

29


Update<br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

Clinical evaluation of lenograstim use in the<br />

Bank of Cyprus <strong>Oncology</strong> Centre<br />

The purpose of this observational study was to evaluate the effect of lenograstim (G-CSF) prophylaxis on febrile<br />

neutropenia incidence in chemotherapy-receiving patients with solid tumours.<br />

Introduction<br />

Febrile neutropenia<br />

Cytotoxic chemotherapy suppresses<br />

the haematopoietic system<br />

impairing host-protective mechanisms.<br />

Neutropenia, the most serious<br />

haematologic toxicity, can lead<br />

to febrile neutropenia (FN), which<br />

usually requires immediate hospitalisation<br />

and antibiotic treatment<br />

[1, 2]. The mean level of in-hospital<br />

mortality associated with<br />

Andri Kouroufexi<br />

MSc<br />

patients hospitalised with FN is 9.5%. This percentage rises above<br />

21% for patients with co-morbidities [3]. Chemotherapy dose<br />

reductions and dose delays, as a result of neutropenia and FN, can<br />

lead to reduced patient survival [4, 5].<br />

The granulocyte-colony stimulating factors<br />

Granulocyte colony-stimulating factors (G-CSFs) stimulate the proliferation<br />

and survival of neutrophils and their precursors, thereby<br />

reducing the incidence and severity of neutropenic complications<br />

across a range of malignancies, and facilitating the delivery of fulldose<br />

chemotherapy [6]. Lenograstim is the glycosylated recombinant<br />

form of human G-CSF. Several randomised controlled trials<br />

have confirmed the efficacy and safety of these agents, and a metaanalysis<br />

involving chemotherapy-receiving adult cancer patients<br />

was recently performed [7]. All forms of G-CSF evoked significant<br />

reductions in FN risk in both solid tumour patients and those with<br />

non-Hodgkin’s lymphoma. Notably, a significant reduction in FN<br />

risk was observed across a broad range of baseline levels of risk<br />

ranging from 17 to 90%. A significant reduction in infection-related<br />

and early all-cause mortality was also demonstrated. These observations<br />

are consistent with that of a Cochrane meta-analysis of therapeutic<br />

CSF in patients hospitalised with FN following cancer<br />

chemotherapy [8]. The meta-analysis also confirmed the ability of<br />

these agents to sustain chemotherapy-relative dose intensity.<br />

Economic considerations<br />

Despite the ability of G-CSFs to reduce the risk of serious<br />

chemotherapy-related toxicities, many patients who receive<br />

myelosuppressive chemotherapy do not receive concomitant<br />

myeloid growth factors. While the decision to utilise a G-CSF in<br />

chemotherapy patients should be based primarily on clinical indications,<br />

the cost of these agents often raises economic considerations<br />

at the institutional and societal level. The direct costs of<br />

myeloid growth factors should be balanced against the reduction in<br />

costs of FN hospitalisation, the reduction in early infection-related<br />

mortality, and overall survival. It is anticipated that by identifying<br />

Stavroula Theophanous-<br />

Kitiri, MSc<br />

George Polykarpou<br />

MSc<br />

patients who are most at risk from<br />

these complications, i.e. those who<br />

are most likely to benefit from prophylactic<br />

myeloid growth factors,<br />

the risk of these serious complications<br />

could be reduced in an efficient<br />

and cost-effective manner.<br />

Clinical practice guidelines<br />

Clinical practice guidelines for the<br />

use of G-CSFs have been developed<br />

by various professional<br />

organisations, including the European Organisation for Research<br />

and Treatment of Cancer (EORTC) [9], the National<br />

Comprehensive Cancer Network [10], and the American Society<br />

of Clinical <strong>Oncology</strong> [11]. They agree that any patient with an FN<br />

risk greater than 20% should receive primary prophylaxis with<br />

G-CSF alongside each chemotherapy cycle. In some instances, the<br />

chemotherapy regimen itself carries an FN risk that exceeds this<br />

threshold. If the FN risk associated with the regimen is 10–20%,<br />

the physician should consider whether patient factors take the<br />

overall risk beyond 20%. If the chemotherapy regimen is considered<br />

to present an FN risk of less than 10%, primary prophylaxis<br />

with G-CSF should not be offered, unless the risk of serious FN<br />

complications is considered high.<br />

Patients and methods<br />

An evaluation of the use of lenograstim in actual practice as measured<br />

by the incidence of subsequent FN in patients with solid<br />

tumours who undergo chemotherapy would be useful for clinicians<br />

who are seeking to develop an evidence-based reimbursement<br />

policy for these drugs.<br />

Consequently, we conducted such a study in the Bank of Cyprus<br />

<strong>Oncology</strong> Centre, Nicosia, Cyprus. A total of 482 patients who<br />

received G-CSF between February 2008 and January 2010 were<br />

identified and selected from the pharmacy computer system Power<br />

Pro. Patient data and blood test results were retrospectively<br />

obtained from medical records and Mosaiq software, a standard<br />

data collection form was developed, and the following information<br />

was recorded: height, weight, date of birth, diagnosis, stage of disease,<br />

and details of prior or concomitant radiotherapy. Any delays<br />

in the administration of chemotherapy (with explanations) were<br />

recorded, as was the absolute neutrophil count at the time of delay,<br />

and details of whether the patient had experienced neutropenia<br />

during the cycle in question. When the dose of administered<br />

chemotherapy was less than optimal, the reason for the dose reduction<br />

and the relative percentage of the administered dose versus the<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

31


planned dose were recorded. Use of G-CSF and antibiotics—<br />

reason, dose and frequency of administration, total number of<br />

doses—were also recorded. Results were recorded in Microsoft<br />

Excel and analysed using an SPSS database.<br />

Results<br />

Between February 2008 and January 2010, 1,791 patients admitted<br />

to the Centre received chemotherapy. Eight hundred and fortysix<br />

(47.2%) patients received lenograstim after myelosuppressive<br />

chemotherapy. Of these 846 lenograstim courses, 482 (57%) were<br />

included in the study, see Figure 1.<br />

Figure 1: Patients receiving lenograstim between February<br />

2008–January 2010<br />

Included in<br />

study<br />

482/846 (57%)<br />

patients<br />

G-CSF<br />

846/1,761 (47.2%)<br />

patients<br />

Lenograstim administration<br />

A total of 12,037 administrations of lenograstim were recorded in<br />

482 patients. The median number of injections administered per<br />

patient per chemotherapy cycle was 5 (range 1–10 injections). In<br />

most patients, administration was initiated either 48 hours (57% of<br />

cases) or 72 hours (36% of cases) post-chemotherapy. Overall,<br />

76.1% (367/482) of patients received prophylactic administration of<br />

lenograstim. The distribution of cancer types was 61.8% (298/482)<br />

breast, 12.4% (60/482) lung, 7.5% (36/482) lymphoma, 3.1%<br />

(15/482) head/neck, 2.7% (13/482) colorectal, 2.7% (13/482) sarcoma,<br />

2.5% (12/482) testicular, 2.5% (12/482) gynaecological, and<br />

4.8% (23/482) other, see Figure 2. The majority of recruited patients<br />

were female (75%), consistent with this tumour-type distribution.<br />

Figure 2: Patient diagnosis<br />

February 2008–January 2010<br />

1,761 patients<br />

Not included<br />

in study<br />

364/846 (43%)<br />

patients<br />

No G-CSF<br />

945/1,761 (52.8%)<br />

patients<br />

Breast<br />

Lung<br />

Lymphoma<br />

Head/Neck<br />

Colorectal<br />

Sarcoma<br />

Testis<br />

Gynaecological<br />

Other<br />

Incidence of FN<br />

Following primary prophylaxis with lenograstim, the incidence of<br />

FN varied between the different cancer types. The malignancies<br />

Update<br />

associated with the highest FN incidence were sarcoma and<br />

head/neck cancer [30.8% (4/13) and 26.7% (4/15), respectively].<br />

Breast cancer was associated with the smallest FN incidence [5%<br />

(15/298)], see Figure 3.<br />

Figure 3: Incidence of FN after lenograstim administration<br />

Delivery of full-dose chemotherapy<br />

Ninety-seven per cent (468/482) of patients received full-dose<br />

chemotherapy as per protocol. Overall, nine courses of chemotherapy<br />

were delayed for ≥ 7 days in 9/482 (2%) patients, and 5/482<br />

(1%) patients required dose reduction ≥ 15% due to prolonged<br />

neutropenia, see Figure 4.<br />

Figure 4: Delivery of full dose chemotherapy<br />

Antibiotic therapy<br />

Ninety-six episodes of FN (82 patients) requiring hospitalisation<br />

occurred while patients were receiving prophylactic G-CSF. The<br />

mean duration of hospital stay was five days. Sarcoma and lung<br />

cancer patients required the longest period of hospitalisation (mean<br />

six days). The empiric antibiotic of choice was piperacillin/tazombactam<br />

+ gentamycin, as administered in 82.4% (79/96) patients.<br />

Ciprofloxacin 500 mg was administered in 73.6% (50/68) patients.<br />

Positive cultures were found only in seven patients (8.5%). Of the<br />

organisms isolated, 5/7 (71.4%) were gram-positive bacteria and<br />

2/7 (28.6%) were gram-negative bacteria.<br />

Adverse events<br />

Various adverse events were observed following primary prophylaxis<br />

with G-CSF. The most frequently reported adverse<br />

event was bone pain, which was recorded in 30.7% (148/482)<br />

of patients. This bone pain was mild to moderate in severity,<br />

and in most patients (83.8%), was successfully treated with<br />

paracetamol.<br />

32 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

FN incidence<br />

patients (%)<br />

Sarcoma<br />

Head/Neck<br />

Testis<br />

Cancer type<br />

Lung<br />

Lymphoma<br />

Breast<br />

Colorectal<br />

Gynaecological<br />

Complete chemotherapy<br />

Dose delay ≥ 7 days<br />

Dose reduction ≥ 15%


Assessment of risk for neutropenic complications according to<br />

EORTC guidelines<br />

According to EORTC guidelines for G-CSF administration, 19.6%<br />

(72/367) of our patients who received primary prophylaxis would<br />

be categorised in the low-risk FN category, 30.8% (113/367)<br />

would be categorised in the intermediate-risk category, and 49.6%<br />

(182/367) would be categorised in the high-risk category. Our lowrisk<br />

patients reported a smaller incidence of FN, but a higher incidence<br />

of neutropenic complications, suggesting that prophylactic<br />

administration of growth factors in these patients improved overall<br />

outcome. Numbers, however, were small, see Figure 5.<br />

Figure 5:Assessment of risk for neutropenic complications<br />

as per EORTC guidelines<br />

< 10% FN risk<br />

72/367 (19.6%)<br />

5/72<br />

(8.3%)<br />

4/72<br />

(5.6%)<br />

2/72<br />

(2.8%)<br />

Primary prophylaxis<br />

67/482 (76.1%)<br />

10–20% FN risk<br />

113/367 (30.8%)<br />

7/113<br />

(6.2%)<br />

2/113<br />

(1.8%)<br />

1/113<br />

(0.8%)<br />

Patients who received<br />

lenograstim<br />

482<br />

> 20% FN risk<br />

182/367<br />

17/182<br />

(9.3%)<br />

3/182<br />

(1.6%)<br />

4/182<br />

(2.2%)<br />

Secondary prophylaxis<br />

115/482 (23.9%)<br />

Febrile<br />

neutropenia<br />

Dose delay ><br />

7 days<br />

Dose reduction<br />

> 15%<br />

Conclusion<br />

The results of our study showed that almost half of the patients who<br />

received chemotherapy in the Centre also received lenograstim.<br />

More than half of these patients were women with breast cancer.<br />

Following lenograstim primary prophylaxis, almost all patients<br />

received full-dose chemotherapy without any modifications.<br />

Our results also showed that G-CSF may currently be misdirected<br />

toward low-risk patients, incurring unnecessary expense.<br />

According to guidelines, G-CSF treatment has limited cost-effectiveness<br />

in low-risk patients, in whom alternative management<br />

approaches are safe and inexpensive. Conversely, in extremely<br />

high-risk populations, it is unlikely that G-CSF therapy could<br />

decrease costs, since early hospital discharge is rarely feasible.<br />

Therefore, it is probably an intermediate-to-high-risk population<br />

that is most likely to benefit from G-CSF therapy, and thus,<br />

lenograstim should be used as part of the standard therapy in the<br />

management of FN patients with solid tumours meeting these risk<br />

criteria.<br />

It is evident that healthcare practitioners should improve G-CSF<br />

use in the hospital setting to facilitate cost-effective therapy. Our<br />

results have assessed many factors, which may lead to modifications<br />

in practice, new guideline recommendations and better outcomes<br />

in the Centre. Perhaps every cancer hospital should undertake<br />

their own assessment and produce their own guidelines for<br />

managing such patients. Further studies are warranted to determine<br />

the best efficacious and cost-effective G-CSF prophylactic options<br />

for patients at each level of risk.<br />

Author for correspondence<br />

Andri Kouroufexi, MSc<br />

Clinical Pharmacist<br />

Makarios Hospital<br />

Nicosia, Cyprus<br />

Co-authors<br />

Stavroula Theophanous-Kitiri 1 , MSc<br />

Clinical Pharmacist<br />

Head of Pharmacy Department<br />

George Polykarpou 1 , MSc<br />

Clinical Pharmacist<br />

Yiola Markou, MD<br />

Medical Oncologist<br />

1 Bank of Cyprus <strong>Oncology</strong> Centre<br />

Strovolos, Cyprus<br />

Evangelia Papadimitriou, PhD<br />

Department of Pharmacy<br />

University of Patras, Greece<br />

References<br />

1. Crawford J, Dale DC, Lyman GH. Chemotherapy-induced neutropenia:<br />

risks, consequences, and new directions for its management. Cancer.<br />

2004;100(2):228-37.<br />

2. Dale DC, McCarter GC, Crawford J, Lyman GH. Myelotoxicity and dose<br />

intensity of chemotherapy: reporting practices from randomized clinical<br />

trials. J Natl Compr Canc Netw. 2003;1(3):440-54.<br />

3. Kuderer NM, Dale DC, Crawford J, Cosler LE, Lyman GH. Mortality,<br />

morbidity, and cost associated with febrile neutropenia in adult cancer<br />

patients. Cancer. 2006;106(10):2258-66.<br />

4. Bosly A, Bron D, Van Hoof A, De Bock R, Berneman Z, Ferrant A, et al.<br />

Achievement of optimal average relative dose intensity and correlation<br />

with survival in diffuse large B-cell lymphoma patients treated with<br />

CHOP. Ann. Hematol. 2008;87(4):277-83.<br />

5. Chirivella I, Bermejo B, Insa A, Perez-Fidalgo A, Magro A, et al. Impact<br />

of chemotherapy dose-related factors on survival in breast cancer patients<br />

treated with adjuvant anthracycline-based chemotherapy. J Clin Oncol.<br />

2006;24(18S):668.<br />

6. Lyman GH, Kuderer NM, Djulbegonic B. Prophylactic granulocyte colony-stimulating<br />

factor in patients receiving dose-intensive cancer chemotherapy:<br />

a meta-analysis. Am J Med. 2002;112(5):406-11.<br />

7. Kuderer NM, Dale DC, Crawford J, Lyman GH. Impact of primary prophylaxis<br />

with granulocyte colony-stimulating factor on febrile neutropenia<br />

and mortality in adult cancer patients receiving chemotherapy: a systematic<br />

review. J Clin Oncol. 2007. 20;25(21):3158-67.<br />

8. Clark OA, Lyman GH, Castro AA, Clark LG, Djulbegovic B. Colony-stimulating<br />

factors for chemotherapy-induced febrile neutropenia: a meta-analysis<br />

of randomized controlled trials. J Clin Oncol. 2005;23(18):4198-214.<br />

9. Aapro MS, Cameron DA, Pettengell R, Bohlius J, Crawford J, Ellis M, et<br />

al. EORTC guidelines for the use of granulocyte-colony stimulating factor<br />

to reduce the incidence of chemotherapy-induced febrile neutropenia<br />

in adult patients with lymphomas and solid tumors. Eur J Cancer.<br />

2006;42(15):2433-53.<br />

10. Crawford J, Althaus B, Armitage J, Balducci L, Bennett C, Blayney DW,<br />

Cataland SR, et al. National Comprehensive Cancer Network (NCCN).<br />

Myeloid growth factors. Clinical practice guidelines in oncology. J Natl<br />

Compr Canc Netw. 2007 Feb;5(2):117.<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

33


ANNOUNCEMENT<br />

European Journal of <strong>Oncology</strong> Pharmacy (EJOP) is the first publication<br />

in oncology pharmacy in the world, and is recognised as the leading<br />

publication in oncology pharmacy in Europe.<br />

Starting from 2007, Volume 1, Issue 1, EJOP has been indexed in<br />

the Directory of Open Access Journals (DOAJ).<br />

DOAJ lists open access journals, that is, scientific and scholarly<br />

journals that meet high quality standards by exercising peer review<br />

or editorial quality control and are free to all from the time of publication<br />

based on the Budapest Open Access Initiative definition of<br />

open access.<br />

Because open access is a worldwide phenomenon, DOAJ includes<br />

publications from around the world in many languages. It is pos-<br />

sible to browse through the journals, or search for articles within<br />

many of the journals through a web interface. In February <strong>2011</strong>, the<br />

database contained 6,100 journals, of which 2,591 were searchable<br />

at article level. In addition, DOAJ participates in the<br />

WorldWideScience global science gateway.<br />

After an evaluation process, DOAJ has decided to index EJOP.<br />

EJOP is honoured to be part of this expert index of research literature.<br />

One can find EJOP easily on www.doaj.org<br />

EJOP is published in its fifth year since its first launch in 2007 and<br />

is also indexed in Embase and Scopus.<br />

To learn more about EJOP, contact editorial@ppme.eu or visit<br />

www.ejop.eu<br />

Cold water reconstitution of Vidaza® with subsequent refrigerated storage prolongs drug stability<br />

References (please see article on pages 24–25)<br />

1. Giralt SA, Horowitz M, Weisdorf D, Cutler<br />

C. Review of stem-cell transplantation for<br />

myelodysplastic syndromes in older<br />

patients in the context of the decision memo<br />

for allogeneic hematopoietic stem cell<br />

transplantation for myelodysplastic syndrome<br />

emanating from the centers for medicare<br />

and medicaid services. J Clin Oncol.<br />

<strong>2011</strong>;29(5):566-72.<br />

2. Beisler JA. Isolation, characterization, and<br />

properties of a labile hydrolysis product of<br />

the antitumor nucleoside, 5-azacytidine. J<br />

Med Chem. 1978; 21(2):204-8.<br />

3. Daher GC, Harris BE, Diasio RB. Meta-<br />

bolism of pyrimidine analogues and their<br />

nucleosides. Pharmacol Ther. 1990;48(2):<br />

189-222.<br />

4. Fenaux P, Mufti GJ, Hellstrom-Lindberg E,<br />

et al. Efficacy of azacitidine compared with<br />

that of conventional care regimens in the<br />

treatment of higher-risk myelodysplastic<br />

syndromes: a randomised, open-label, phase<br />

III study. Lancet Oncol. 2009;10:223-32.<br />

5. ICH Guidance Q2 (R1). Validation of analytical<br />

procedures: text and methodology.<br />

November 2005. Available from: http://www.<br />

ich.org/fileadmin/ Public_Web_Site/ICH_<br />

Products/Guidelines/Quality/Q2_R1/Step4/<br />

Q2_R1_Guideline.pdf<br />

6. ICH Guidance Q6 A. Specifications: test<br />

procedures and acceptance criteria for new<br />

drug substances and new drug products: chemical<br />

substances. October 1999. Available<br />

from: http://www.ich.org/fileadmin/Public_<br />

Web_Site/ICH_ Products/Guidelines/Quality/<br />

Q6A/Step4/Q6Astep4.pdf<br />

7. ICH Guidance Q1 A (R2). Stability testing<br />

of new drug substances and products.<br />

February 2003. Available from: http://www.<br />

ich.org/fileadmin/Public_Web_Site/ICH_<br />

Products/Guidelines/Quality/Q1A_R2/Step4<br />

/Q1A_R2_Guideline.pdf<br />

34 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu


Special <strong>Report</strong><br />

For personal use only. Not to be reproduced without permission of the publisher (copyright@ppme.eu).<br />

Hospital workers perceptions about nanotechnology<br />

Nagwa Ibrahim, PharmD, FAIHQ; Ali Al Zahrani, MD, FRCR; Ashraf Al Alwan, PhD<br />

Nanotechnology is the tiny world of controlling matter at the scale of one billionth of an atom. In the last<br />

few years, nanotechnology has catapulted from being the specialty of a few physicists and chemists to a<br />

worldwide scientific and industrial enterprise.<br />

Introduction<br />

The rapid development of nanotechnology and the growing importance<br />

of this technology for everyday life have not really attracted<br />

any attention from the public at large. However, little is known<br />

about the technology’s possible health and environmental implications.<br />

At this critical juncture, it is important that leaders from<br />

industry, government, the science and engineering community, and<br />

other sectors develop a better understanding of what the public<br />

wants and expects of these new and emerging technologies [1-3].<br />

The number of projects that encourage the public to engage with<br />

nanotechnology is growing all the time. However, some scientists<br />

are uncomfortable with the idea that non-experts should have<br />

active roles in decisions about nanotechnology, because they are<br />

often uninformed. Regardless of this opinion, public engagement<br />

is becoming a serious component of nanotechnology policy in<br />

many countries [1-6]. Public perception will have a major influence<br />

on the success of new applications of nanotechnology, as will<br />

the results of risk assessments carried out by industry on the various<br />

nanotechnology applications [1, 5-8].<br />

Objective<br />

The aim of this study was to evaluate hospital employee perception<br />

about nanotechnology in Saudi Arabia and correlate it with<br />

existing demographic data.<br />

Methodology<br />

Three hundred surveys were distributed to employees and trainees<br />

from different departments in our hospital. The response rate to the<br />

survey was 40% (120/300). Thirty-nine surveys (33%) were<br />

excluded because less than 70% of the questionnaire was complet-<br />

ed. The survey included nine core questions which measured hospital<br />

workers awareness, perceptions and preferences of nanotechnology.<br />

Demographic measures, such as age, sex, profession, years<br />

of experience and level of education, were also recorded.<br />

Results<br />

Demographics<br />

Table 1 shows that we managed to achieve a relatively representative<br />

distribution of men and women in our sample, with 51% of<br />

respondents being female. The highest percentage of participants<br />

(29%) were aged between 30–40 years and the lowest percentage<br />

(9%) were aged between 50–60 years. At least 68% of respondents<br />

had achieved a bachelor’s degree and 21% had a doctorate or<br />

equivalent level. A slight majority of respondents (52%) were<br />

pharmacists, whilst 21% were physicians. Thirty-five per cent of<br />

respondents had less than five years’ experience.<br />

By correlating age with whether workers had heard about nanotechnology,<br />

we observed a similarity between the group aged<br />

between 18–25 years and the group aged between 30–40 years.<br />

Sixteen per cent of each of these two groups reported that they had<br />

heard of nanotechnology, whilst 11% did not. Six per cent of the<br />

groups aged 25–30 years and 40–50 years stated that they had not<br />

heard of nanotechnology, whilst 10% and 14% respectively, had.<br />

In the 50–60 years age group, 5% had and 5% had not.<br />

With regards to education, the highest percentage of employees<br />

who had heard of nanotechnology were those with a doctorate or<br />

equivalent degree. This was followed by university graduates, university<br />

undergraduates, and those with a Masters degree (19%,<br />

15%, 12%, and 9% respectively).<br />

Table 1: Demographic data (N = 81)<br />

N = 81<br />

Age 18–25 25–30 30–40 40–50 50–60<br />

21 (26%) 13 (16%) 23 (29%) 16 (20%) 7 (9%)<br />

Sex Male Female N/A<br />

34 (42%) 41 (51%) 6<br />

Level of education Doctorate Master University<br />

graduate<br />

University<br />

undergraduate<br />

Diploma or<br />

high school<br />

17 (21%) 10 (13%) 27 (34%) 16 (19%) 11 (13%)<br />

Profession Medicine Pharmacy Nursing Others<br />

17 (21%) 42 (52%) 12 (15%) 10 (12%)<br />

Years of experience < 5 5–10 10–15 15–20 > 20<br />

28 (35%) 16 (20%) 14 (17%) 7 (8%) 10 (12%)<br />

N: total number of participants; N/A: not applicable.<br />

European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu<br />

35


Table 2: Questions and answers<br />

In terms of experience, the highest percentage of employees that<br />

had heard of nanotechnology was those with less than five years of<br />

experience. This was followed by those with 10–15 years, those<br />

with 5–10 years, those with over 20 years, and those with 15–20<br />

years (17%, 12%, 11%, 7%, and 5% respectively).<br />

Survey question answers<br />

Table 2 shows that in general, 61% of people in Saudi Arabia reported<br />

that they had heard of nanotechnology. The largest proportion of<br />

respondents (40%) indicated that they heard a little about nanotechnology.<br />

The majority of the respondents (40%) had heard about nanotechnology<br />

through the media, while the remaining proportion<br />

knew about nanotechnology from conferences, Internet, books, and<br />

other sources respectively. A slim majority of respondents (51%)<br />

agreed with the statement, ‘Overall, I support the use of nanotechnology<br />

for human enhancement’. Thirty-eight per cent agreed that<br />

the benefits of nanotechnology outweighed its risks, 15% agreed that<br />

its benefits and risks were equal, and 27 % were not sure about the<br />

risk and benefits of nanotechnology. The majority (47%) indicated<br />

that they considered that nanotechnology would be very important<br />

to science and business in the next five years, 21% considered that it<br />

would be fairly important, and 15% were unsure. Almost 75% indicated<br />

that they required more information about nanotechnology.<br />

The majority of respondents also tended to associate nanotechnology<br />

usage equally within medicine and pharmacy (67% and 65%<br />

of respondents respectively).<br />

Conclusion<br />

Anticipatory governance of emergent technologies depends on a<br />

comprehensive understanding of the values in society that shape<br />

public understanding and opinion of new and emerging technolo-<br />

Special <strong>Report</strong><br />

Have you heard about Yes No<br />

nanotechnology? 50 (61%) 31 (39%)<br />

How much have you A lot Some A little Nothing Not sure<br />

heard, read or seen about 5 (7%) 18 (22%) 32 (40%) 11 (14%) 3 (4%)<br />

using nanotechnology?<br />

How did you know about Conference Media Internet Books Others<br />

nanotechnology? 20 (25%) 32 (40%) 21 (26%) 5 (6%) 6 (7%)<br />

I support use of nano- Agree Slightly agree Do not know Slightly disagree Disagree<br />

technology for human 41 (51%) 9 (11%) 16 (20%) Not applicable 1 (2%)<br />

enhancement<br />

What is your initial Benefits Benefits and risks Risks outweigh Not sure<br />

impression of risks and outweigh risks are equal benefits<br />

benefits of nanotechnology? 31 (38%) 12 (15%) Not applicable 22 (27%)<br />

Rate the impression of Very important Fairly important Not too Not at all Do not know<br />

nanotechnology to science important important<br />

and business in the next 38 (47%) 17 (21%) Not applicable Not applicable 12 (15%)<br />

five years<br />

Do you feel that you need Yes No Not interested<br />

to know more information 61 (75%) Not applicable 5 (6%)<br />

about nanotechnology?<br />

Do you recommend to Yes No Do not know<br />

policy makers to provide 49 (61%) Not applicable 16 (20%)<br />

more information to<br />

hospital workers about<br />

nanotechnology?<br />

gies, and of related current technologies. This survey aimed to<br />

measure and evaluate the understanding of public values about nanotechnology<br />

in Saudi Arabia, as presented by hospital workers.<br />

Public opinion research should be considered a vital element in any<br />

attempt to assess a developing technology, and in any subsequent<br />

efforts at anticipatory governance. Further research is highly recommended.<br />

In addition, we recommend that policy makers should<br />

increase hospital employee awareness about nanotechnology<br />

through continuous scientific events. This may lead to improved<br />

understanding and indirect future advances in nanotechnology. We<br />

plan to re-evaluate and assess the workers’perceptions in five years<br />

time. A comparison of available data with the international public<br />

perception about nanotechnology is also recommended.<br />

Authors<br />

Nagwa Ibrahim, PharmD, FAIHQ<br />

Clinical Pharmacist Specialist<br />

Pharmacy Department<br />

Ali Al Zahrani, MD, FRCR<br />

Director, Consultant<br />

<strong>Oncology</strong> Department<br />

Ashraf Al Alwan, PhD<br />

Assistant Director, Consultant Clinical Pharmacist<br />

Clinical Services, Pharmacy Department<br />

Riyadh Military Hospital<br />

PO Box 7897<br />

SA-11159 Riyadh, Saudi Arabia<br />

References<br />

References can be found on page 17.<br />

36 European Journal of <strong>Oncology</strong> Pharmacy • Volume 5 • <strong>2011</strong> • Issues 3-4 www.ejop.eu

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!